EP0813606B1 - Method of purification of recombinant viral vectors containing a therapeutic gene - Google Patents

Method of purification of recombinant viral vectors containing a therapeutic gene Download PDF

Info

Publication number
EP0813606B1
EP0813606B1 EP96911282A EP96911282A EP0813606B1 EP 0813606 B1 EP0813606 B1 EP 0813606B1 EP 96911282 A EP96911282 A EP 96911282A EP 96911282 A EP96911282 A EP 96911282A EP 0813606 B1 EP0813606 B1 EP 0813606B1
Authority
EP
European Patent Office
Prior art keywords
resin
purification
deae
acn53
lysate
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Lifetime
Application number
EP96911282A
Other languages
German (de)
French (fr)
Other versions
EP0813606A2 (en
Inventor
Paul W. Shabram
Bernard G. Huyghe
Xiaodong Liu
H. Michael Shepard
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Canji Inc
Original Assignee
Canji Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Canji Inc filed Critical Canji Inc
Priority to EP06075526A priority Critical patent/EP1686180A3/en
Publication of EP0813606A2 publication Critical patent/EP0813606A2/en
Application granted granted Critical
Publication of EP0813606B1 publication Critical patent/EP0813606B1/en
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/82Translation products from oncogenes
    • CCHEMISTRY; METALLURGY
    • C08ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
    • C08FMACROMOLECULAR COMPOUNDS OBTAINED BY REACTIONS ONLY INVOLVING CARBON-TO-CARBON UNSATURATED BONDS
    • C08F265/00Macromolecular compounds obtained by polymerising monomers on to polymers of unsaturated monocarboxylic acids or derivatives thereof as defined in group C08F20/00
    • C08F265/02Macromolecular compounds obtained by polymerising monomers on to polymers of unsaturated monocarboxylic acids or derivatives thereof as defined in group C08F20/00 on to polymers of acids, salts or anhydrides
    • CCHEMISTRY; METALLURGY
    • C09DYES; PAINTS; POLISHES; NATURAL RESINS; ADHESIVES; COMPOSITIONS NOT OTHERWISE PROVIDED FOR; APPLICATIONS OF MATERIALS NOT OTHERWISE PROVIDED FOR
    • C09DCOATING COMPOSITIONS, e.g. PAINTS, VARNISHES OR LACQUERS; FILLING PASTES; CHEMICAL PAINT OR INK REMOVERS; INKS; CORRECTING FLUIDS; WOODSTAINS; PASTES OR SOLIDS FOR COLOURING OR PRINTING; USE OF MATERIALS THEREFOR
    • C09D11/00Inks
    • C09D11/02Printing inks
    • C09D11/10Printing inks based on artificial resins
    • C09D11/106Printing inks based on artificial resins containing macromolecular compounds obtained by reactions only involving carbon-to-carbon unsaturated bonds
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/05Animals comprising random inserted nucleic acids (transgenic)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/10011Adenoviridae
    • C12N2710/10311Mastadenovirus, e.g. human or simian adenoviruses
    • C12N2710/10341Use of virus, viral particle or viral elements as a vector
    • C12N2710/10343Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/10011Adenoviridae
    • C12N2710/10311Mastadenovirus, e.g. human or simian adenoviruses
    • C12N2710/10351Methods of production or purification of viral material
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S435/00Chemistry: molecular biology and microbiology
    • Y10S435/803Physical recovery methods, e.g. chromatography, grinding

Definitions

  • Size exclusion chromatography for purification of various plant viruses has been demonstrated either as a stand alone technique or to augment density gradient centrifugation (Albrechtsen et al. , J. Virological Methods 28:245-256 (1990); and Hewish et al., J. Virological Methods 7:223-228 (1983)). Size exclusion appears promising for bovine papilloma virus (Hjorth and Mereno-Lopez, J. Virological Methods 5:151-158 (1982)); and has been shown to be a superior method for the purification of tick-borne encephalitis virus (Crooks et al., J. Chrom. 502:59-68 (1990)).
  • Adenoviruses are large (diameter of approximately 80 nm) and somewhat fragile.
  • a large literature base dealing with the relationship of structure to function has accumulated (for reviews see Philipson, Virology 15:263-268 (1961) and Horwitz, Virology (Second Edition) Raven Press Ltd, New York (1990)). Little has been reported in the literature about chromatographic purification of live adenoviruses. Haruna et al.
  • Virology 13:264-267 (1961) reported using DEAE ion exchange chromatography for purification of types 1, 3 and 8 adenoviruses while Klemperer and Pereir ( Virology 9:536-545 1959)) and Philipson ( Virology 10:459-465 (1960)) reported difficulties using the same method with other types of adenoviruses.
  • WO9502688 reported the purification of viral vectors using sulfated oligosaccharides columns. Poor resolution and poor yield are important problems with this methodology that has prevented its use in large-scale production.
  • the chromatographically purified viral vector containing the therapeutic gene is as pure and active as a viral vector purified using a three day ultracentrifugation.
  • the purification method of this invention provides several advantages over existing methods including quality of purified viral vector, consistency, decreased process time and the ability to process large amounts of sample. Furthermore, because this novel purification scheme is based on the surface characteristics of the virion it is broadly applicable to other virions using the teaching of this invention as well as to virions containing different internal DNA constructs with different therapeutic genes. This break-through purification method is an important aspect in bulk commercialization of gene therapy.
  • the invention provides a method of purifying viral vectors containing therapeutic genes for use in gene therapy.
  • the invention comprises a method of purification from a cell lysate of a recombinant viral vector containing a therapeutic gene, which comprises: a) treating said lysate with an enzymatic agent that selectively degrades both unencapsulated DNA and RNA; b) chromatographing the treated lysate from step a) on a first resin; and c) chromatographing the eluant from step b) on a second resin; wherein one resin is an anion exchange resin and the other is an immobilized metal ion chromatography (IMAC) resin.
  • IMAC immobilized metal ion chromatography
  • hydrophobic interaction chromatography resin may be substituted for the immobilized metal ion chromatography resin.
  • the enzymatically treated cell lysate undergoes a filtration step adding a fourth step to the three step method.
  • the viral vector being purified is an adenoviral vector containing a therapeutic gene.
  • the adenoviral vector is a recombinant type 5 adenovirus with an internal DNA construct including a tumor suppressor gene.
  • This invention is directed to a method for purification of a recombinant viral vector containing a therapeutic gene from a cell lysate, which comprises:
  • viral vector means a recombinant virus that has had some or all of the genes in the native genome removed such that the virus is replication- incompetent.
  • the viral vector shall mean viruses wherein the recombinant viral genome contains a DNA encoding a therapeutic gene such that the viral vector is used to transfer the therapeutic gene to a desired human cell for gene therapy.
  • Representative vectors include those that will infect mammalian, and especially human, cells, and are derived from viruses chosen from the group consisting of retroviruses, adenoviruses, herpes viruses and avipox viruses.
  • Retroviral and adenoviral vectors are preferred. Adenoviral vectors, especially type 2 and type 5 adenoviral vectors, are especially preferred. The recombinant type 5 adenoviral vectors are the most preferred.
  • therapeutic gene means genes or functional fractions thereof encoding molecules which have a desired therapeutic effect. For example, a gene which either by its absence or mutation causes an increase in pathological cell growth or proliferation of cells. A therapeutic gene as used herein would replace such an absent or mutated gene. Therapeutic genes may give rise to their therapeutic effect either by remaining extrachromosomal such that the gene will be expressed by the cell from the extrachromosomal location or the gene may be incorporated into the genome of the cell such that it recombines with the endogenous gene.
  • Such genes include tumor- suppressor genes, including those chosen from the group consisting of Rb, Rb mutants, p53, p53 mutants, DCC, NF-1, Wilm's tumor, NM 23, BRCA-1, BRCA-2, BRUSH-1, p56, H-NUC, thyroid hormone receptor gene, retenoic acid receptor gene, genes encoding p130, p107, and p85.
  • Other gene replacement or supplementation strategies include adenosine deaminase (ADA), thymidine kinase (TK), genes encoding various cytokines such as y-interferon, ⁇ -interferon, IL-2 and other hormones.
  • Therapeutic gene is also understood to include DNA encoding a ribozyme.
  • DNA constructs encode an RNA enzyme which binds and destroys the RNA of selected positively-acting growth regulatory genes such as (1) oncogenes or proto-oncogenes selected from the group consisting of, but not limited to, the following: c-myc, c-fos, c-jun, c-myb, c-ras, Kc and JE; (2) growth factor receptor genes selected from the group consisting of, but not limited to, epidermal growth factor, platelet derived growth factor, transferrin and insulin.
  • Preferred therapeutic genes are the tumor suppressor genes, with most preferred tumor suppressor genes being Rb, Rb mutants, p53, p53 mutants, BRUSH-1, p56, BRCA-1, BRCA-2, p16 and p21.
  • cell lysate means a collection of cells, including host cells which contain a vector, preferably a viral vector, which cells have been removed from their growth environment and have had their cell membranes disrupted by physiological or chemical means.
  • enzymatic agent means a compound or mixture that selectively degrades unencapsulated DNA and RNA, while not disrupting the recombinant viral vectors to the extent they are not infectious and are unable deliver an intact copy of the therapeutic gene to the desired target cell.
  • Such an enzymatic agent is generally a combination of one or more enzymes referred to as endonucleases or DNAses or RNAses.
  • BenzonaseTM is a preferred enzymatic reagent.
  • anion exchange resin shall mean a positively-charged organic moiety covalently cross-linked to an inert polymeric support.
  • Representative organic moieties are drawn from primary, secondary, tertiary and quaternary amino groups; such as trimethylaminoethyl (TMAE), diethylaminoethyl (DEAE), dimethylaminoethyl (DMAE), and other groups such as the polyethyleneimine (PEI) that already have, or will have, a formal positive charge within the pH range of approximately 5 to approximately 9.
  • representative negatively changed organic groups are chosen from the group consisting of carboxymethyl and sulfomethyl and other groups that have, or will have, a formal negative charge within the pH range of about 5 to about 9
  • the support material should be one that is easily derivatizable and possess good mechanical strength.
  • the material can be a natural polymeric substance, a synthetic polymer or co-polymer, or a mixture of natural and synthetics polymers.
  • the support can take the shape of porous or nonporous particles, beads, membranes, disks or sheets.
  • Such supports include silica, hydrophilic polymer (MonoBeads®, Pharmacia, Piscataway, New Jersey), cross-linked cellulose (e.g. Sephacel®), cross-linked dextran (e.g. Sephadex®) cross-linked agarose (e.g.
  • Sepharose® polystyrene
  • a co-polymer such as polystyrene-divinylbenzene or one composed of oligoethyleneglycol, glycidylmethacrylate and pentaerythroldimethacrylate, to which are grafted polymerized chains of acrylamide derivatives (the latter co-polymer is known as a "tentacle” support).
  • the resins can be used in a traditional (gravity) column chromatography or high pressure liquid chromatography apparatus using radial or axial flow, fluidized bed columns, or in a slurry (batch) method. In the latter method, the resin is separated from the sample by decanting or centrifugation or filtration or a combination of methods.
  • the viral vectors can be purified then eluted from these resins by an increasing salt gradient, preferably a gradient of sodium chloride.
  • anion exchange resins examples include Fractogel® (E. Merck, Gibbstown, New Jersey) resins derivatized with either DEAE or DMAE; Fractogel® EMD Tentacle resins derivatized with DEAE, DMAE, or TMAE; Toyopearl® (TasoHaas, Montgomeryville, Pennsylvania) resins derivatized with DEAE or QAE; Acti-Disk® (Whatman, Clifton, New Jersey) supports derivatized with Quat, DEAE or PEl; Sepharose® (Pharmacia, Piscataway, New Jersey) resins derivatized with DEAE; Sephacel® (Pharmacia, Piscataway, New Jersey) resins derivatized with DEAE; and Sephadex® resins derivatized with DEAE and QAE.
  • Preferred anion exchange resins are derivatized with the DEAE group, and further preferred columns are the Fractogel, Toyopeart and StreamlineTM (
  • IMMAC immobilized metal ion affinity chromatography
  • the metal chelating groups a are those known in the art to bind to zinc. nickel, copper, cobalt, calcium or magnesium ions in the formal (+2) oxidation state.
  • Such groups include the imminodiacetic (IDA) group, the tris(carboxymethyl)ethylenediamine (TED) groupip, the N-(hydroxyethyl)ethylenediaminetriacetic group, and derivatives suruch as the N-(methyl), and the N-(hydroxymethyl) IDA groups.
  • the chelalating groups can be bound to any of the above-listed metals, with zinc the preferred metal.
  • the description of the chemical composition, physical form and u uses of the polymeric supports described above for the term "anion exchangage resin" applies to the IMAC resin as well.
  • Cross-linked agarose and the "a tentacle" supports are preferred.
  • the viral vectors can be eluted from the b IMAC column by adding increasing concentrations of competing chelatisting agents such as imidazole, histamine, glycine, or ammonium chloride, or, alternatively the pH of the eluant can be raised or lowered, as long as the extremes of the range of the ph gradient used remain from about 5 to about 9.
  • competing chelatisting agents such as imidazole, histamine, glycine, or ammonium chloride
  • Examples of commercially-available products that can be used in the instant method include the Acti-Disk IDA cartridge (Whatman), Fractogel AF chelate, and Toyopearl AF chelate IMAC resins.
  • the preferred conditions for the immobilized metal affinity ion resin for the purification of the viral vectors such as type 5-adenovirus derived vector on DEAE had the resin charged with a divalent metal cation chosen from the group consisting of cobalt, nickel, copper, zinc, calcium and magnesium and further wherein the resin is an IDA or TED cross-linked agarose resin, especially an IDA agarose resin that is charged with zinc ion.
  • a hydrophobic interaction chromtagraphy can be substituted for the IMAC resin in the instant method.
  • Such a resin has lower alkyl or aryl groups covalently bound through a non-polar group, such as an aliphatic ether, to an inert polymeric support.
  • Lower alkyl groups such as methyl, propyl, n-butyl, neo-pentyl, and octyl and the phenyl group are examples of the interactive group on the instant resin.
  • Butyl is the preferred interactive group.
  • Cross-linked agarose, hydroxylated polyether, hydrophilic media and silica are the preferred supports with cross-linked agarose the most preferred.
  • the viral vectors can be purified then eluted from the HIC resin by a decreasing salt gradient, with ammonium sulfate the preferred salt.
  • Examples of commercially available HIC columns useful for the current invention include the cross-linked agarose columns such as Phenyl-, Butyl and Octyl Sepharose, and Toyopearl (Phenyl, Propyl, and Butyl) and Fractogel (Propyl or Phenyl).
  • the IMAC or HIC resin can be used before or after the anion exchange resin. If used before, the salt concentration of the eluant from the HIC or IMAC resin should diluted to about 450 millimolar or less in order to prevent premature stripping of viral particles from the exchange resin.
  • buffer or “buffered solution” refers to a mixture of acid and base which when present in a solution reduces or modulates changes in pH that would otherwise occur in the solution when acid or based is added.
  • the cell lysate is maintained in a buffered solution. Suitable buffers are those that can maintain the pH of the resultant solution between about 5.0 and about 9. 0.
  • buffers are commercially available and include phosphate, MES, HEPES, MOPS, Borate, TRIS, BES, ADA, ACES, PIPES, MOPSO, BIS-TRIS PROPANE, BES, TES, DIPSO, TAPSO, TRIZMA, HEPPSO, POPSO, TEA, EPPS, TRICINE, GLYCYLGLYCINE, BICINE, TAPS, and the like.
  • Preferred buffers include the phosphate, MES, HEPES, MOPS, borate, and TRIS, with HEPES being the most preferred.
  • chromatography for the purification of viral vectors, such as an adenoviral vector referred to as Type 5, for use in gene therapy has been shown to be an effective alternative to cesium chloride density gradient ultracentrifugations.
  • the purification scheme described in this invention selects for product based on the surface characteristics of the virion. These characteristics do not change with different internal DNA constructs, e.g. having different therapeutic genes in the construct, therefore leading to similar chromatographic behaviors. In other words, the chromatography is unaffected by the nature of the therapeutic gene inserted inside the vector.
  • the anion exchange resins immobilized metal ion affinity chroromatography resins and hydrophobic interaction chromatography resins are cleaned using methods known to the ordinarily skilled arartisan.
  • DEAE is treated first with NaOH then HCl and finnally with NaCl.
  • the IZAC is first treated with EDTA then NAOH. HCL, NaClCl flushing with H 2 O in between each step.
  • the columns are equilibrated in apappropriate buffers using appropriate binding conditions. Columns are loaded d with sample in a buffer such that the product will bind to the resin by cocontrolling pH and salt concentration for DEAE and by controlling pH, salt concerentration and divalent metal ion concentration for the immobilized metal ion affininity column. The columns are washed to remove contaminants and may be reuused.
  • inventions include the additioronal step of filtering the cell lysate after it is treated with the enzymatic agent I
  • the cell lysate is buffered before treatment with Benzonase at a pH between about 5.0 and about 9.0 before applyiying it to the first resin.
  • Benzonase at a pH between about 5.0 and about 9.0 before applyiying it to the first resin.
  • These embodiments are preferred for purifying a recombinant viral vector derived from either a retrovirus or a adenovirus, and especially so when the vector is derived from an adenonvirus, and such a vectoror contains a tumor suppressor gene.
  • adenoviral-derived vectoror containing a tumor suppressor gene are those when the treated, buffered cell ill lysate is first chromatographed over an anion exchange resin followed by chromatography over a immobilized metal affinity resin. These coronditions are preferred especially when the recombinant viral vector is derived infrom either a type 2 or type 5 adenovirus, and especially a type 5 adenovirus.
  • the enzymatic agent used to treat the cell lysate is one or morene enzymes, especially those chosen from the group consisting of RNAse and D DNAse or a mixture of endonucleases as would be known to the ordinarily skillelled artisan.
  • the preferred enzymatic agent for use in this embodiment is BenzonaseTM, a recombinant non-specific nuclease which cleaves both RNA and DNA.
  • an especially preferred method of the one described in the preceding paragraphs is where the type 5 adenoviral-derived recombinant viral vector has a genome containing the wild-type p53 gene.
  • Standard recombinant Adenovirus Type 5 designated ACN53 is a vector derived from a Type 5 Adenovirus which has the F1 coding sequence with a 1.4-kb full length p53 cDNA driven by the human cytomegalovirus promoter (Wills et al., Human Gene Therapy 5:1079-1088 (1994)).
  • Virus was prepared by a 3-step centrifugation procedure as described (Laver et al., Virology 45:598-614 (1971)) with the following modifications. Infected cells were lysed by 3 cycles of freeze-thaw and centrifuged at 15,000 rpm for 10 min, 4°C in a Sorvall RC-5B centrifuge.
  • the pellet was discarded, and the supernatant was treated with BenzonaseTM (American International Chemical, Natick, Mass.) at 133 U/mL for 30 min at room temperature.
  • the treated material was then layered onto a 1.25 gm/mL and 1.40 gm/mL CsCl discontinuous step gradient in 10 mM Tris pH8.1 and centrifuged at 30,000 rpm for 75 min, 10°C in a Sorvall TST 41-14 rotor.
  • the virus band from each tube was collected, pooled, mixed with 1.35 gm/mL CsCl (in 10mM Tris pH8.1) and centrifuged overnight at 45,000 rpm in a Beckman VTi 50 rotor at 10°C.
  • the virus band from each tube was collected and recentrifuged at 45,000 rpm as before for an additional 4 hrs.
  • the final virus pool from this step was dialyzed extensively against phosphate buffered saline (PBS) supplemented with 2% sucrose and 2 mM MgCl 2 at a temperature of 4°C.
  • PBS phosphate buffered saline
  • the purified virus was used to infect human embryonic kidney 293 cells (available from American Type Culture Collection), as described below in Procedure 2.
  • ATCC 293 ATCC CRL 15763 cells were grown in a Cell FactoryTM (Nunc, Ruskilde, Denmark) in a CO 2 incubator in 1.5L of medium consisting of DME high glucose medium containing 10% Hyclone bovine serum defined supplemented, 2 mM glutamine (Irvine Scientific, Santa Ana, California), 1 mM sodium pyruvate (Irvine Scientific). No antibiotics were added to the medium.
  • the cells were infected with a multiplicity of infection (MOI) of 5 to 10 infectious units per cell in 500 mL of fresh medium. The virus was added to the medium, mixed thoroughly, and introduced to the cells in the unit.
  • MOI multiplicity of infection
  • the membrane was incubated with the primary antibody (Cytimmune rabbit IgG a- adenovirus type 5, Lee Biotechnology Research: San Diego, CA) at 5 ⁇ g/mL (in TBS) for 1 hr at room temperature. Following primary incubation, the membrane was washed 3 times with TBS and incubated with the secondary antibody (Amersham Life Sciences, Arlington Heights, IL) Horseradish peroxidase conjugated anti-rabbit lg) diluted to 1 ⁇ L stock antibody/1 mL TBS for 1 hr at room temperature.
  • the primary antibody Cytimmune rabbit IgG a- adenovirus type 5, Lee Biotechnology Research: San Diego, CA
  • the secondary antibody Amersham Life Sciences, Arlington Heights, IL
  • concentrated virus was diluted 1 in 10 in 0.1% SDS in phosphate buffered saline (PBS).
  • PBS phosphate buffered saline
  • the sample was vortexed for 1 minute and then centrifuged at 14,000 rpm in an Ependorf microfuge to remove any precipitate.
  • a matched pair of cuvettes were blanked with 0.1 % SDS in PBS buffer by running a baseline scan on Shimadzu UV160U spectrophotometer (Shimadzu Scientific Instrument, Columbia, MD.).
  • the SDS-treated virus sample was then placed in the sample cuvette and scanned from 220 to 340 nm. If there was absorbance between 310 and 320 nm, the sample was too concentrated and was diluted further and remeasured.
  • the A 260 /A 280 ratio was also determined from this scan, and must be between 1.2-1.3 in order to ensure that the product is pure enough to calculate particle number. If this condition is met, the absorbance value at 260 nm only is used to calculate the number of virions/mL. A conversion factor of 1.1 x 10 12 particles per absorbance unit at 260 nm (Maizel et al., Virology 36:115-125 (1968)) was used to calculate particle number with approximately 10% error. Typical values for samples subjected only to Anion Exchange Chromotography (i.e., Examples 1 and 2 using a DEAE resin) were between 1.14 to 1.19. When such samples were subjected to an IZAC as described in Example 3, the ratios were in the 1.22 to 1.25 range.
  • Infected cell lysate is comprised of contaminants both host-cell and viral in origin. Some of these contaminants were removed prior to chromatography. Specifically, host cell, non encapsulated or incomplete ACN53 nucleic acids were enzymatically degraded at this stage of the process with a nuclease such as BenzonaseTM. This may be done early in the process for two reasons. Early BenzonaseTM treatment resulted in better yields in anion exchange chromatography. BenzonaseTM was removed by subsequent process steps. The presence of Benzonase during the process can be assayed by a commercially available ELISA kit (American International Chemical, Natick, MD.)
  • Clarification of the treated lysate was accomplished by filtration rather than centrifugation. A slow speed spin was used to remove cellular debris (Fig. 1). Filtration was then used to prepare the product for loading onto the first column. The type of filter used (i.e. composition and pore size) and its effective product recovery was assayed by the quantitative anion exchange assay described in Example 3, Procedure A.
  • Centrifugation was followed by filtration through a Gelman Sciences AcrodiscTM 0.8/0.2 ⁇ m 2-stage syringe filter. Recovery of ACN53 depended on the pore size and type of membrane used for filtration. Filters such as polysulfone, PVDF membranes and cellulose acetate based membranes were used. Polysulfone and PVDF were preferred.
  • the supernatent from this step was made 2 mM in MgCl 2 , 2% (wt/vol) in sucrose and 2.5% (wt/vol) ⁇ -cyclodextrin.
  • BenzonaseTM (American International Chemical, Inc.) was added to a final concentration of 100 units/mL and allowed to incubate for 1 hr at room temperature.
  • the treated material was clarified by centrifugation in a Beckman TJ-6 at 3000 rpm for 10 min and filtration through a Gelman Sciences AcrodiscTM 0.8/0.2 ⁇ m filter. The resultant supernatant was taken on to Example 2.
  • the pH can be varied, however, if run at pH 7.5 using HEPES pH 7.5/NaCl/sucrose/MgCl 2 less contaminating material bound to the columns.
  • DEAE chromatography yielded a high degree of initial purification.
  • the immobilized metal ion purification chromatography step removed these contaminants.
  • Anion exchange resins were equilibrated in 50 mM HEPES, pH 7.5, 300 mM NaCl, 2 mM MgCl 2 , 2% sucrose at 1 mL/min.
  • a 50 mM Tris buffer pH 8.0 (with 300 mM NaCl, 2 mM MgCl 2 , 2% sucrose) was also used.
  • elution was performed with a 20 column volume 300-600 mM linear NaCl gradient and collected in 0.5 mL fractions. In preparation for future use, the column was then cleaned with 2 column volumes of 0.5 M NaOH, a 1.5 M NaCl wash and re-equilibrated.
  • CsCl-ACN53 was injected onto a DEAE column equilibrated in 50mM Tris pH 8 at 2 mL/min (350 cm/hr) and eluted with a 10 min (11.7 column volume) 0-1.5 M linear NaCl gradient. A single peak was detected with an A 260 /A 280 of 1.23. The protein bands present in this fraction reacted with Ad 5 polyclonal antibody upon slot-blot analysis.
  • peaks were resolved when an infected cell lysate sample was applied to the DEAE column (Fig. 2).
  • the composition of the peaks were deduced from the A 260 /A 280 absorbance ratio.
  • the first peak has an A 260 /A 280 ratio of 0.5, and is mainly protein.
  • the third peak has an A 260 /A 280 ratio of 2, suggesting that this material was nucleic acid.
  • the ACN53 virus peak eluted second with a ratio of 1.23.
  • the identities of these peaks were confirmed by spiking experiments and by running SDS gels of each peak. In typical experiments using the above conditions an A 260 /A 280 ratio of 1.23 ⁇ .08 was found to be characteristic of virus peaks.
  • Immobilized metal affinity chromatography (IMAC) using zinc as the divalent cation (IZAC) gave higher product recovery and did not require any sample manipulation of the DEAE fraction pool prior to loading. Impurities removed by this method eluted in the flow through peak and were well resolved from product, leading to simpler pooling criteria.
  • IZAC was reproducible, and when used with DEAE provided a two column purification protocol capable of delivering pure ACN53 as specified by SDS-PAGE gels and westerns, A 260 A 280 ratios and infectious to non-infectious particle ratios.
  • the Immobilized zinc affinity chromatography (IZAC) system was prepared for metal charging by washing the column sequentially with 1 volume of 100 mM EDTA and 1 volume of 0.2 M NaOH, flushing with water after each step.
  • the matrix was subsequently charged with zinc by injecting 1 column volume of 100 mM ZnCl 2 in H 2 0 acidified with 0.5 ⁇ L/mL glacial acetic acid.
  • the matrix was then thoroughly washed in water prior to equilibration in 50 mM HEPES pH 7.5/450 mM NaCl/2% sucrose/2 mM MgCl 2 .
  • Sample loading did not require any manipulation; DEAE pool fractions or CsCl derived material could be injected directly onto the column.
  • the column was washed with a 10 column volume decreasing NaCl linear gradient from 50 mM HEPES pH 7.5/450 mM NaCl/2% sucrose/2 mM MgCl 2 to 50 mM HEPES pH 7.5/150 mM NaCl/2% sucrose/2 mM MgCl 2 . Elution was performed with a linear 0-500 mM glycine gradient (in 150 mM NaCl) applied over 10 column volumes.
  • ACN53-DEAE from Example 2 fraction pool purified over IZAC is shown in Fig.5.
  • a gel and western blot comparison of CsCl-ACN53, DEAE purified and DEAE/IZAC purified material can be seen in Figs 6 and 7.
  • the CsCl-ACN53 and DEAE/IZAC materials were very similar, and the DEAE-only purified material was less pure by these criteria.
  • IZAC buffer and elution systems were evaluated by splitting a DEAE-ACN53 pool in half and purifying both halves over IZAC in HEPES pH 7.5 and Tris pH 8 buffer systems. IZAC was also run in the presence of sucrose and MgCl 2 without affecting the separation. The use of copper as the metal ion and imidazole as the elution agent were also tested (for a general review of metal affinity chromatography, see Belew et.al. 1987; Kato et.al, 1986). Various systems: zinc/glycine, zinc/imidazole, copper/glycine and copper/imidazole, all worked on the metal affinity purification columns. ACN53 can be eluted using a pH 7 to pH9 gradient. A : Assay for Recombinant Adenoviral Particle Number by Anion Exchange HPLC.
  • a 1 mL Resource Q- (Pharmacia, Piscataway, NJ) anion exchange column was used to quantitate the number of viral particles in various samples.
  • the column was equilibrated in 300 mM NaCl, 50 mM HEPES, pH 7.5. at a flow rate of 1 mL /min on a Waters 625 chromatography system equipped with 717plus autosampler and a 991 photodiode array detector (PDA). The chromatography was monitored on the PDA detector (Milford, MA) scanning from 210 to 300 nm.
  • a standard curve was constructed by injecting CsCl purified ACN53 virions characterized for total particles at a selected absorbance, A 260 in 0.1%SDS.
  • the assay was independent of injected sample volume. After the sample loaded, the column was washed with two column volumes of equilibration buffer followed by a linear gradient from 300 to 600 mM NaCl in 50 mM HEPES, pH 7.5, over 10 column volumes. The gradient was followed with a 2 column volume wash with 600 mM NaCl in 50mM HEPES, pH 7.5. After each chromatographic run, the column was cleaned with 2.6 column volumes of 1.5 M NaCl in 50 mM HEPES, pH 7.5, and then re-equilibrated for the next injection. The column was cleaned more vigorously after injection of crude samples by injecting 0.25 to 1 column volume of 0.5 N NaOH followed by a wash with 1.5 M salt.
  • a variety of buffer conditions have been used in the purification of ACN53 by column chromatography in this study in the pH range of 7.0-8.0. (e.g. Seth Virology 68:1204-1206 (1994)). Checking for degradation at various pH limits is accomplished by assaying for degradation by TCID50 (Procedure C below) and analytical anion exchange analysis (Procedure A above). The effect of buffer salt concentration on the viability of ACN53 showed that abrupt changes of ionic strength can lead to loss of virus. Salt concentration should be carefully controlled and monitored.
  • ATCC 293 cells were plated into a 96-well plate: 100 ⁇ L of 5 x 10 5 cells/mL for each well in complete MEM (10% bovine calf serum; 1% glutamine) media.
  • complete MEM 10% bovine calf serum; 1% glutamine
  • a 250 ⁇ L aliquot of virus sample diluted 1:10 6 was added to the first column and is serially diluted two-fold across the plate.
  • One row was reserved for a positive control (CsCl-purified ACN53); one for a negative control.
  • a 100 ⁇ L aliqout of each well was transferred to its identical position in the ATCC-293 seeded plate and allowed to incubate a 37° in a humidified incubator for 2 days.
  • the ratio of total virus particles to infectious viral particles can vary widely from preparation to preparation. Values for CsCl derived viruses in the range of 60 to 80. The calculation of this ratio in crude lysates or semi-pure fractions has been made possible by the anion exchange particle assay (see Example 3[A]). Using analytical anion exchange changes in the ratio of ideal virus particles to infectious virus particles can be monitored. Using the purification method of this invention the total virus particle to infectious virus particle ratio of the crude lysate and subsequently purified material is comparable to that obtained using ultracentrafugation. Within the error of the assay these values are equivalent. By this criteria, chromatographic purification is well tolerated by the virus. By other criteria, namely SDS-PAGE and western blot analysis (Figs. 6 and 7), chromatographic purification is superior to ultracentrifugation based methods.
  • Saos-2 cells a p53-negative osteosarcoma cell line.
  • Saos-2 cells were seeded into a 6-well tissue culture plate at a concentration of 5 x 10 5 cells/well in 3 mL of media: Kaighn's nutrient mixture
  • F12 DME High Glucose (1:1 mixture), supplemented with 2 mM L-glutamine and 10% fetal bovine serum.
  • the cells were incubated in humidified air, 7 ⁇ 1% CO 2 at 37° for 16-24 hrs.
  • the spent media were removed and replaced with 1 mL of fresh media, and the cells were infected at 20,40, or 60 MOI of purified virus.
  • Host cell contamination was assessed by western blot analysis using polyclonal antibodies developed against ATCC-293 cell components. Polyclonal antibodies were commercially obtained and were raised against various 293 cellular antigens (HTI, Ramona, CA). The results indicated that the final products of either CsCl or DEAE-IZAC purification contained no detectable host cell contaminants. In the case of semi-pure DEAE-ACN53, there was a small amount of immunogenic contamination seen in the product pool: 3 major bands and several minor ones. The majority of host cell contamination is recovered in the flow-through portion of the DEAE step. Contaminants which copurified with ACN53 in the DEAE step were removed by zinc affinity chromatography, and were recovered in IZAC flow-through fractions prior to the introduction of the glycine gradient.
  • Gels to be silver stained were loaded with 5-15 ⁇ L of sample.
  • the sample was boiled with an equal volume of reducing buffer and electrophoresed as described for Coomassie detection. Fixing was performed by treating the gel in 10% trichloroacetic acid for 1 hr followed by a 3x wash in water purified to 18 megohms.
  • the gel was stained with the Daiichi silver staining kit according to the instructions provided (Integrated Separation Systems).
  • a preferred purification scheme for ACN53 as shown above is to treat infected cell lysate with BenzonaseTM prior to the chromatographic steps. Clarification is then accomplished by step filtration through 0.8 ⁇ m followed by 0.2 ⁇ m membranes. If necessary, a larger pore (e.g. 5 ⁇ m) prefiltration step can be added for more viscous suspensions. Adjustment to pH 7.5/300 mM NaCl is then performed in preparation for loading onto a DEAE column. The product peak, as detected by the A 260 /A 280 nm ratio or the characteristic photo-diode array spectrum, is pooled and directly injected onto a zinc-charged, iso-osmotically equilibrated metal affinity column.
  • the ionic strength of the buffer is then gradually lowered to approximate phosphate-buffered saline (ca,150 mM NaCl) prior to elution of product with a glycine gradient. This material is then dialyzed into the final formulation.

Abstract

The invention provides a method for purifying viral vectors containing therapeutic genes for use in gene therapy. The invention comprises a method of purification from a cell lysate of a recombinant viral vector containing a therapeutic gene, which comprises: a) treating said lysate with an enzymatic agent that selectively degrades both unencapsulated DNA and RNA; b) chromatographing the treated lysate from step a) on a first resin; and c) chromatographing the eluant from step b) on a second resin; wherein one resin is an anion exchange resin and the other is an immobilized metal ion chromatography (IMAC) resin.

Description

    Background of the Invention
  • The treatment of disease by gene therapy has moved from the theoretical to the practical realm. The first human gene therapy trial was begun in September 1990 and involved transfer of adenosine deaminase (ADA) gene into lymphocytes of patients with defects of this enzyme. Lack of ADA activity results in immune deficiency. Of several methods for delivering therapeutic genes to diseased cells, viral vectors hold out particular promise. Tumor suppressor genes are being investigated for treating cancerous cells. Viral vectors containing such tumor suppressor genes are being evaluated as potential therapeutics in the field of cancer therapy. Recently, adenovirus vectors have received particular attention as an advantageous vector for the delivery of such tumor suppressor genes and other biological response modifiers. As studies of cancer gene therapy progress to clinical trials, larger and larger quantities of purified viral vectors are needed. One problem in producing suitable quantities of vectors for such trials is the purification of the particles from the cell lysates in which the viral particles are produced.
  • Specifically, purification of these vectors has historically been performed by using density-based ultracentrifugation methods. While this method has proven effective for use as a research tool, it is not feasible as a method for industrial scale production. Meeting the demands for material in the future would lead to prohibitive costs unless a new purification scheme could be developed. One alternative to ultracentrifugation is chromatographic techniques for purification of infectious viral particles.
  • The use of size exclusion chromatography for purification of various plant viruses has been demonstrated either as a stand alone technique or to augment density gradient centrifugation (Albrechtsen et al., J. Virological Methods 28:245-256 (1990); and Hewish et al., J. Virological Methods 7:223-228 (1983)). Size exclusion appears promising for bovine papilloma virus (Hjorth and Mereno-Lopez, J. Virological Methods 5:151-158 (1982)); and has been shown to be a superior method for the purification of tick-borne encephalitis virus (Crooks et al., J. Chrom. 502:59-68 (1990)). The use of size exclusion chromatography has not yet become widespread, but is currently being employed for large scale production of recombinant retroviruses (Mento, S. J., Viagene, Inc. as reported at the 1994 Williamsburg Bioprocessing Conference). Affinity chromatography, mostly using monoclonal antibodies (Mab), has been reported to be an effective method for the purification of antigens of viral origin (Najayou et al., J. Virological Methods, 32:67-77. 1991). Soybean mosaic virus (a virus which can survive pH 3) can be recovered using Mab affinity chromatography (Diaco et al., J. Gen. Virol. 67:345-351. 1986). Fowler (J. Virological Methods. 11:59-74. (1985)) used affinity chromatography and density gradient centrifugation to purify Epstein Barr virus.
  • Adenoviruses are large (diameter of approximately 80 nm) and somewhat fragile. A large literature base dealing with the relationship of structure to function has accumulated (for reviews see Philipson, Virology 15:263-268 (1961) and Horwitz, Virology (Second Edition) Raven Press Ltd, New York (1990)). Little has been reported in the literature about chromatographic purification of live adenoviruses. Haruna et al. (Virology 13:264-267 (1961)) reported using DEAE ion exchange chromatography for purification of types 1, 3 and 8 adenoviruses while Klemperer and Pereir (Virology 9:536-545 1959)) and Philipson (Virology 10:459-465 (1960)) reported difficulties using the same method with other types of adenoviruses. WO9502688 reported the purification of viral vectors using sulfated oligosaccharides columns. Poor resolution and poor yield are important problems with this methodology that has prevented its use in large-scale production.
  • Thus, a need exists for a chromatographic method for purifying viral vectors such as adenovirus vectors.
  • Summary of the Invention
  • One obstacle to the successful practice of gene therapy is the availability of purified viral vectors to deliver therapeutic genes. The present invention solves that problem by the unexpected and surprising discovery that viral vectors containing therapeutic genes can be purified sufficiently for therapeutic and/or prophylactic use using a three step process comprising: enzymatically treating the cell lysate comprising the viral vector containing the therapeutic gene; chromatographing the enzymatically treated cell lysate on a first resin; and chromatographing the eluate from the first column on a second resin. The resulting purified viral vectors having the therapeutic gene retained their infectivity during and after chromatographic treatment and are able to effect gene transfer. For the first time it was found using the purification method of this invention that the chromatographically purified viral vector containing the therapeutic gene is as pure and active as a viral vector purified using a three day ultracentrifugation. The purification method of this invention provides several advantages over existing methods including quality of purified viral vector, consistency, decreased process time and the ability to process large amounts of sample. Furthermore, because this novel purification scheme is based on the surface characteristics of the virion it is broadly applicable to other virions using the teaching of this invention as well as to virions containing different internal DNA constructs with different therapeutic genes. This break-through purification method is an important aspect in bulk commercialization of gene therapy.
  • Therefore, the invention provides a method of purifying viral vectors containing therapeutic genes for use in gene therapy. In one embodiment, the invention comprises a method of purification from a cell lysate of a recombinant viral vector containing a therapeutic gene, which comprises: a) treating said lysate with an enzymatic agent that selectively degrades both unencapsulated DNA and RNA; b) chromatographing the treated lysate from step a) on a first resin; and c) chromatographing the eluant from step b) on a second resin; wherein one resin is an anion exchange resin and the other is an immobilized metal ion chromatography (IMAC) resin.
  • In an alternative embodiment a hydrophobic interaction chromatography resin may be substituted for the immobilized metal ion chromatography resin.
  • In an alternative embodiment the enzymatically treated cell lysate undergoes a filtration step adding a fourth step to the three step method.
  • In a more preferred embodiment the viral vector being purified is an adenoviral vector containing a therapeutic gene. In a still more preferred embodiment the adenoviral vector is a recombinant type 5 adenovirus with an internal DNA construct including a tumor suppressor gene.
  • Brief Description of the Figures
    • Figure 1: Recovery of ACN53 from the supernatant of centrifuged crude infected cell lysate which was centrifuged for 5 minutes at different speeds in an Eppendorf centrifuge model 5415c at 4°. Analysis for ACN53 was by analytical anion exchange.
    • Figure 2: Identification of ACN53 infected cell lysate components and separated by DEAE chromatography by dual wavelength UV absorbance using their 260/280 nm absorbance ratio.
    • Figure 3: Comparison of CsCI-ACN53 and host-cell contaminant retention times during DEAE purification. Elution of CsCI-ACN53 is compared to elution of uninfected H293 host-cell lysate blank chromatographed over a DEAE column.
    • Figure 4: Butyl-Hydrophobic Interaction Chromatography of a DEAE-ACN53 fraction pool. A semi-pure DEAE purified ACN53 fraction pool was mixed with an equal volume of 50 mM tris/pH 8.0/3 M ammonium sulfate and injected onto a TosoHaas Butyl-650M column and eluted with a 1.5 - 0 M decreasing ammonium sulfate gradient.
    • Figure 5: Immobilized metal affinity chromatography of a DEAE-ACN53 fraction pool. A semi-pure DEAE purified ACN53 fraction pool was injected onto a 6.6 x 50 mm TosoHaas AF chelate 650M column charged with ZnCl2 and eluted with a linear 0-500 mM glycine gradient.
    • Figure 6: SDS-PAGE comparison of ACN53 derived from column chromatography and CsCI-ultracentrifugation. Samples were electrophoresed on an 8-16% gradient polyacrylamide gel and silver stained for analysis. Lanes 2 and 3 are DEAE and IZAC eluate pools respectively. Lanes 4-6 represent 3 different lots (CsCl-1, CsCl-2, and CsCl-3) of CsCl-ACN53 run side by side in order to examine lot to lot consistency. Lane 7, CsCl-ion exchange treated lot, represents the ACN53 peak recovered when a sample of Cscl-ACN53 derived ion-exchange treated lot is purified over a Resource Q anion exchange HPLC column (Resource Q, Pharmacia). Lanes 1 and 8 are standard molecular weight markers.
    • Figure 7: Western Blot Comparison of ACN53 Derived from Column Chromatography and CsCl-Ultracentrifugation. Samples, identical to those previously described, were electrophoresed on an 8-16% gradient gel and transferred to a PVDF membrane. The blot was incubated first with 5 µg/ml Cytimmune rabbit IgG anti adenovirus type 5 antibody, then with Amersham's horseradish peroxidase conjugated anti-rabbit lg (NA934) and developed using electrochemical detection.
    • Figure 8: Expression of p53 gene product in Saos-2 cells. Two different lots of chromatographically produced ACN53 were assayed by western blot for their ability to affect gene transfer to p53-null Saos-2 cells. The semi-pure DEAE fractions are shown in lanes 7 & 8, the final product in 5 & 6. P53-expressing SW480 cells were used as a positive control; uninfected Saos-2 cells were used as a negative control.
    Detailed Description of the Invention
  • This invention is directed to a method for purification of a recombinant viral vector containing a therapeutic gene from a cell lysate, which comprises:
  • a) treating the cell lysate with an enzymatic agent that selectively degrades both unencapsulated DNA and RNA; b) chromatographing the treated lysate from step a) on a first resin; and c) chromatographing the eluant from step b) on a second resin; wherein one such resin is an anion exchange resin and the other is an immobilized metal ion affinity resin.
  • The term "viral vector" means a recombinant virus that has had some or all of the genes in the native genome removed such that the virus is replication- incompetent. Furthermore, the viral vector shall mean viruses wherein the recombinant viral genome contains a DNA encoding a therapeutic gene such that the viral vector is used to transfer the therapeutic gene to a desired human cell for gene therapy. Representative vectors include those that will infect mammalian, and especially human, cells, and are derived from viruses chosen from the group consisting of retroviruses, adenoviruses, herpes viruses and avipox viruses. Retroviral and adenoviral vectors are preferred. Adenoviral vectors, especially type 2 and type 5 adenoviral vectors, are especially preferred. The recombinant type 5 adenoviral vectors are the most preferred.
  • The term "therapeutic gene" means genes or functional fractions thereof encoding molecules which have a desired therapeutic effect. For example, a gene which either by its absence or mutation causes an increase in pathological cell growth or proliferation of cells. A therapeutic gene as used herein would replace such an absent or mutated gene. Therapeutic genes may give rise to their therapeutic effect either by remaining extrachromosomal such that the gene will be expressed by the cell from the extrachromosomal location or the gene may be incorporated into the genome of the cell such that it recombines with the endogenous gene. Such genes include tumor- suppressor genes, including those chosen from the group consisting of Rb, Rb mutants, p53, p53 mutants, DCC, NF-1, Wilm's tumor, NM 23, BRCA-1, BRCA-2, BRUSH-1, p56, H-NUC, thyroid hormone receptor gene, retenoic acid receptor gene, genes encoding p130, p107, and p85. Other gene replacement or supplementation strategies include adenosine deaminase (ADA), thymidine kinase (TK), genes encoding various cytokines such as y-interferon, α-interferon, IL-2 and other hormones.
  • Therapeutic gene is also understood to include DNA encoding a ribozyme. Such DNA constructs encode an RNA enzyme which binds and destroys the RNA of selected positively-acting growth regulatory genes such as (1) oncogenes or proto-oncogenes selected from the group consisting of, but not limited to, the following: c-myc, c-fos, c-jun, c-myb, c-ras, Kc and JE; (2) growth factor receptor genes selected from the group consisting of, but not limited to, epidermal growth factor, platelet derived growth factor, transferrin and insulin.
  • Preferred therapeutic genes are the tumor suppressor genes, with most preferred tumor suppressor genes being Rb, Rb mutants, p53, p53 mutants, BRUSH-1, p56, BRCA-1, BRCA-2, p16 and p21.
  • The term "cell lysate" means a collection of cells, including host cells which contain a vector, preferably a viral vector, which cells have been removed from their growth environment and have had their cell membranes disrupted by physiological or chemical means.
  • The term "enzymatic agent" means a compound or mixture that selectively degrades unencapsulated DNA and RNA, while not disrupting the recombinant viral vectors to the extent they are not infectious and are unable deliver an intact copy of the therapeutic gene to the desired target cell. Such an enzymatic agent is generally a combination of one or more enzymes referred to as endonucleases or DNAses or RNAses. Benzonase™ is a preferred enzymatic reagent.
  • The term "anion exchange resin" shall mean a positively-charged organic moiety covalently cross-linked to an inert polymeric support. Representative organic moieties are drawn from primary, secondary, tertiary and quaternary amino groups; such as trimethylaminoethyl (TMAE), diethylaminoethyl (DEAE), dimethylaminoethyl (DMAE), and other groups such as the polyethyleneimine (PEI) that already have, or will have, a formal positive charge within the pH range of approximately 5 to approximately 9.
  • Similarly, representative negatively changed organic groups are chosen from the group consisting of carboxymethyl and sulfomethyl and other groups that have, or will have, a formal negative charge within the pH range of about 5 to about 9
  • The support material should be one that is easily derivatizable and possess good mechanical strength. The material can be a natural polymeric substance, a synthetic polymer or co-polymer, or a mixture of natural and synthetics polymers. The support can take the shape of porous or nonporous particles, beads, membranes, disks or sheets. Such supports include silica, hydrophilic polymer (MonoBeads®, Pharmacia, Piscataway, New Jersey), cross-linked cellulose (e.g. Sephacel®), cross-linked dextran (e.g. Sephadex®) cross-linked agarose (e.g. Sepharose®), polystyrene, or a co-polymer such as polystyrene-divinylbenzene or one composed of oligoethyleneglycol, glycidylmethacrylate and pentaerythroldimethacrylate, to which are grafted polymerized chains of acrylamide derivatives (the latter co-polymer is known as a "tentacle" support).
  • The resins can be used in a traditional (gravity) column chromatography or high pressure liquid chromatography apparatus using radial or axial flow, fluidized bed columns, or in a slurry (batch) method. In the latter method, the resin is separated from the sample by decanting or centrifugation or filtration or a combination of methods. The viral vectors can be purified then eluted from these resins by an increasing salt gradient, preferably a gradient of sodium chloride.
  • Examples of suitable anion exchange resins include Fractogel® (E. Merck, Gibbstown, New Jersey) resins derivatized with either DEAE or DMAE; Fractogel® EMD Tentacle resins derivatized with DEAE, DMAE, or TMAE; Toyopearl® (TasoHaas, Montgomeryville, Pennsylvania) resins derivatized with DEAE or QAE; Acti-Disk® (Whatman, Clifton, New Jersey) supports derivatized with Quat, DEAE or PEl; Sepharose® (Pharmacia, Piscataway, New Jersey) resins derivatized with DEAE; Sephacel® (Pharmacia, Piscataway, New Jersey) resins derivatized with DEAE; and Sephadex® resins derivatized with DEAE and QAE. Preferred anion exchange resins are derivatized with the DEAE group, and further preferred columns are the Fractogel, Toyopeart and Streamline™ (Pharmacia, Placateway, A New Jersey) DEAE resins.
  • The term "Immobilized metal ion affinity chromatography" ("IMMAC") resin refers to an inert natural or synthetic polymeric support covalently ty cross-linked with a metal chelating group. The metal chelating groups a are those known in the art to bind to zinc. nickel, copper, cobalt, calcium or magnesium ions in the formal (+2) oxidation state. Such groups include the imminodiacetic (IDA) group, the tris(carboxymethyl)ethylenediamine (TED) groupip, the N-(hydroxyethyl)ethylenediaminetriacetic group, and derivatives suruch as the N-(methyl), and the N-(hydroxymethyl) IDA groups. These groups cacan be cross-linked to the natural or synthetic polymeric support by standard alaliphatic ether linkages and reagents, such as bisoxirane, epichlorhydrin, and 1,4 ,4-bis-(2,3-epoxypropoxy)butane. For the method of the invention, the chelalating groups can be bound to any of the above-listed metals, with zinc the preferred metal. The description of the chemical composition, physical form and u uses of the polymeric supports described above for the term "anion exchangage resin" applies to the IMAC resin as well. Cross-linked agarose and the "a tentacle" supports are preferred. The viral vectors can be eluted from the b IMAC column by adding increasing concentrations of competing chelatisting agents such as imidazole, histamine, glycine, or ammonium chloride, or, alternatively the pH of the eluant can be raised or lowered, as long as the extremes of the range of the ph gradient used remain from about 5 to about 9. Examples of commercially-available products that can be used in the instant method include the Acti-Disk IDA cartridge (Whatman), Fractogel AF chelate, and Toyopearl AF chelate IMAC resins.
  • Furthermore, the preferred conditions for the immobilized metal affinity ion resin for the purification of the viral vectors such as type 5-adenovirus derived vector on DEAE had the resin charged with a divalent metal cation chosen from the group consisting of cobalt, nickel, copper, zinc, calcium and magnesium and further wherein the resin is an IDA or TED cross-linked agarose resin, especially an IDA agarose resin that is charged with zinc ion.
  • A hydrophobic interaction chromtagraphy ("HIC") can be substituted for the IMAC resin in the instant method. Such a resin has lower alkyl or aryl groups covalently bound through a non-polar group, such as an aliphatic ether, to an inert polymeric support. Lower alkyl groups such as methyl, propyl, n-butyl, neo-pentyl, and octyl and the phenyl group are examples of the interactive group on the instant resin. Butyl is the preferred interactive group. The chemical composition, physical form and method of use for the supports described In the definition of the term "anion-exchange resin" above also apply to the HIC resins. Cross-linked agarose, hydroxylated polyether, hydrophilic media and silica are the preferred supports with cross-linked agarose the most preferred. The viral vectors can be purified then eluted from the HIC resin by a decreasing salt gradient, with ammonium sulfate the preferred salt. Examples of commercially available HIC columns useful for the current invention include the cross-linked agarose columns such as Phenyl-, Butyl and Octyl Sepharose, and Toyopearl (Phenyl, Propyl, and Butyl) and Fractogel (Propyl or Phenyl).
  • As mentioned above, in the instant invention the IMAC or HIC resin can be used before or after the anion exchange resin. If used before, the salt concentration of the eluant from the HIC or IMAC resin should diluted to about 450 millimolar or less in order to prevent premature stripping of viral particles from the exchange resin.
  • The term "buffer" or "buffered solution" refers to a mixture of acid and base which when present in a solution reduces or modulates changes in pH that would otherwise occur in the solution when acid or based is added. In one embodiment, the cell lysate is maintained in a buffered solution. Suitable buffers are those that can maintain the pH of the resultant solution between about 5.0 and about 9. 0. Such buffers are commercially available and include phosphate, MES, HEPES, MOPS, Borate, TRIS, BES, ADA, ACES, PIPES, MOPSO, BIS-TRIS PROPANE, BES, TES, DIPSO, TAPSO, TRIZMA, HEPPSO, POPSO, TEA, EPPS, TRICINE, GLYCYLGLYCINE, BICINE, TAPS, and the like. Preferred buffers include the phosphate, MES, HEPES, MOPS, borate, and TRIS, with HEPES being the most preferred.
  • The use of chromatography for the purification of viral vectors, such as an adenoviral vector referred to as Type 5, for use in gene therapy has been shown to be an effective alternative to cesium chloride density gradient ultracentrifugations. There are several advantages related to this methodology, including quality, consistency, decreased process time, system automation, and the ability to process large amounts of crude lysate. The purification scheme described in this invention selects for product based on the surface characteristics of the virion. These characteristics do not change with different internal DNA constructs, e.g. having different therapeutic genes in the construct, therefore leading to similar chromatographic behaviors. In other words, the chromatography is unaffected by the nature of the therapeutic gene inserted inside the vector.
  • The anion exchange resins immobilized metal ion affinity chroromatography resins and hydrophobic interaction chromatography resins are cleaned using methods known to the ordinarily skilled arartisan. By way of example, DEAE is treated first with NaOH then HCl and finnally with NaCl. The IZAC is first treated with EDTA then NAOH. HCL, NaClCl flushing with H2O in between each step. The columns are equilibrated in apappropriate buffers using appropriate binding conditions. Columns are loaded d with sample in a buffer such that the product will bind to the resin by cocontrolling pH and salt concentration for DEAE and by controlling pH, salt concerentration and divalent metal ion concentration for the immobilized metal ion affininity column. The columns are washed to remove contaminants and may be reuused.
  • Other embodiments of the instant invention include the additioronal step of filtering the cell lysate after it is treated with the enzymatic agent I In an alternative embodiment the cell lysate is buffered before treatment with Benzonase at a pH between about 5.0 and about 9.0 before applyiying it to the first resin. These embodiments are preferred for purifying a recombinant viral vector derived from either a retrovirus or a adenovirus, and especially so when the vector is derived from an adenonvirus, and such a vectoror contains a tumor suppressor gene.
  • Further embodiments for purifying an adenoviral-derived vectoror containing a tumor suppressor gene are those when the treated, buffered cell ill lysate is first chromatographed over an anion exchange resin followed by chromatography over a immobilized metal affinity resin. These coronditions are preferred especially when the recombinant viral vector is derived infrom either a type 2 or type 5 adenovirus, and especially a type 5 adenovirus.
  • The enzymatic agent used to treat the cell lysate is one or morene enzymes, especially those chosen from the group consisting of RNAse and D DNAse or a mixture of endonucleases as would be known to the ordinarily skillelled artisan. The preferred enzymatic agent for use in this embodiment is Benzonase™, a recombinant non-specific nuclease which cleaves both RNA and DNA.
  • Finally, an especially preferred method of the one described in the preceding paragraphs is where the type 5 adenoviral-derived recombinant viral vector has a genome containing the wild-type p53 gene.
  • Experimental Section Procedure 1 Preparation of ACN53 Standard Material (CsCl-ACN53)
  • Standard recombinant Adenovirus Type 5 designated ACN53 is a vector derived from a Type 5 Adenovirus which has the F1 coding sequence with a 1.4-kb full length p53 cDNA driven by the human cytomegalovirus promoter (Wills et al., Human Gene Therapy 5:1079-1088 (1994)). Virus was prepared by a 3-step centrifugation procedure as described (Laver et al., Virology 45:598-614 (1971)) with the following modifications. Infected cells were lysed by 3 cycles of freeze-thaw and centrifuged at 15,000 rpm for 10 min, 4°C in a Sorvall RC-5B centrifuge. The pellet was discarded, and the supernatant was treated with Benzonase™ (American International Chemical, Natick, Mass.) at 133 U/mL for 30 min at room temperature. The treated material was then layered onto a 1.25 gm/mL and 1.40 gm/mL CsCl discontinuous step gradient in 10 mM Tris pH8.1 and centrifuged at 30,000 rpm for 75 min, 10°C in a Sorvall TST 41-14 rotor. The virus band from each tube was collected, pooled, mixed with 1.35 gm/mL CsCl (in 10mM Tris pH8.1) and centrifuged overnight at 45,000 rpm in a Beckman VTi 50 rotor at 10°C. The virus band from each tube was collected and recentrifuged at 45,000 rpm as before for an additional 4 hrs. The final virus pool from this step was dialyzed extensively against phosphate buffered saline (PBS) supplemented with 2% sucrose and 2 mM MgCl2 at a temperature of 4°C. The purified virus was used to infect human embryonic kidney 293 cells (available from American Type Culture Collection), as described below in Procedure 2.
  • Procedure 2 Production of Infected ATCC 293 Cells, Harvest and Lysis
  • ATCC 293 (ATCC CRL 1573) cells were grown in a Cell Factory™ (Nunc, Ruskilde, Denmark) in a CO2 incubator in 1.5L of medium consisting of DME high glucose medium containing 10% Hyclone bovine serum defined supplemented, 2 mM glutamine (Irvine Scientific, Santa Ana, California), 1 mM sodium pyruvate (Irvine Scientific). No antibiotics were added to the medium.
  • Two to two and a half days after seeding the Cell Factory™, when cell monolayers reached about 50-60% confluency, the cells were infected with a multiplicity of infection (MOI) of 5 to 10 infectious units per cell in 500 mL of fresh medium. The virus was added to the medium, mixed thoroughly, and introduced to the cells in the unit.
  • When cell monolayers from Preparation 1 showed signs of detachment from the surface of the Cell Factory™ (usually at 3 to 4 days post-infection), the cells were harvested by gentle tapping and were centrifuged in a Beckman TJ-6 at 1500 rpm for 5 min. They were washed once with serum free media, pelleted again, and resuspended in 25 mL of 50mM Tris buffer pH 8.0/150 mM NaCl, 2 mM MgCl2, 2% sucrose for use in the preparation of ultracentrifuge-derived standard virus. Samples destined for use in the procedures of Examples 1 through 3 were resuspended in 25 mL of 50mM HEPES buffer pH 7.5/150 mM NaCl, 2 mM MgCl2, 2% sucrose. The cells were lysed at this point by 3 cycles of freeze-thaw. Following the third cycle, cellular debris was removed by centrifugation in a Beckman TJ-6 at 1500 rpm for 5 min. At a temperature of 4°C.
  • Procedure 3 Western Blot Analysis of Samples Containing Recombinant Adenovirus Particles.
  • An SDS-PAGE gel was run as described in Example 3, Procedure F with approximately the same loading as that of a silver stained gel. The bands were then transferred to a PVDF membrane pre-wetted in 100% methanol and equilibrated in Tris-buffered saline (TBS). The gel was also equilibrated in TBS. The proteins were transferred to the membrane using a Bio-Rad semi-dry transfer apparatus at 25 V for 30 minutes. The membrane was then blocked in 1% casein/0.01 % sodium azide overnight at 4° or at room temperature for 1 hr, and washed 3 times with TBS. The membrane was incubated with the primary antibody (Cytimmune rabbit IgG a- adenovirus type 5, Lee Biotechnology Research: San Diego, CA) at 5 µg/mL (in TBS) for 1 hr at room temperature. Following primary incubation, the membrane was washed 3 times with TBS and incubated with the secondary antibody (Amersham Life Sciences, Arlington Heights, IL) Horseradish peroxidase conjugated anti-rabbit lg) diluted to 1 µL stock antibody/1 mL TBS for 1 hr at room temperature. A final three time wash was performed with TBS and the membrane incubated with Amersham ECL detection reagents for 1 minute, exposed in the dark to Hyperfilm-ECL (Amersham) for various times (several seconds to minutes to give a selection of various contrasts) and developed in an X-ray film developer.
  • Using this western blot analysis, differences between banding patterns of ACN53 in various states of purification can be seen in Figure 7.
  • Procedure 4 Assay for Recombinant Adenoviral Particle Number by Absorbance at 260nm in Presence of SDS.
  • For this measurement, concentrated virus was diluted 1 in 10 in 0.1% SDS in phosphate buffered saline (PBS). The sample was vortexed for 1 minute and then centrifuged at 14,000 rpm in an Ependorf microfuge to remove any precipitate. A matched pair of cuvettes were blanked with 0.1 % SDS in PBS buffer by running a baseline scan on Shimadzu UV160U spectrophotometer (Shimadzu Scientific Instrument, Columbia, MD.). The SDS-treated virus sample was then placed in the sample cuvette and scanned from 220 to 340 nm. If there was absorbance between 310 and 320 nm, the sample was too concentrated and was diluted further and remeasured. The A260/A280 ratio was also determined from this scan, and must be between 1.2-1.3 in order to ensure that the product is pure enough to calculate particle number. If this condition is met, the absorbance value at 260 nm only is used to calculate the number of virions/mL. A conversion factor of 1.1 x 1012 particles per absorbance unit at 260 nm (Maizel et al., Virology 36:115-125 (1968)) was used to calculate particle number with approximately 10% error. Typical values for samples subjected only to Anion Exchange Chromotography (i.e., Examples 1 and 2 using a DEAE resin) were between 1.14 to 1.19. When such samples were subjected to an IZAC as described in Example 3, the ratios were in the 1.22 to 1.25 range.
  • Example 1 Lysis of Unencapulated Nucleic Acids Nuclease Treatment
  • Infected cell lysate is comprised of contaminants both host-cell and viral in origin. Some of these contaminants were removed prior to chromatography. Specifically, host cell, non encapsulated or incomplete ACN53 nucleic acids were enzymatically degraded at this stage of the process with a nuclease such as Benzonase™. This may be done early in the process for two reasons. Early Benzonase™ treatment resulted in better yields in anion exchange chromatography. Benzonase™ was removed by subsequent process steps. The presence of Benzonase during the process can be assayed by a commercially available ELISA kit (American International Chemical, Natick, MD.)
  • Clarification of the treated lysate was accomplished by filtration rather than centrifugation. A slow speed spin was used to remove cellular debris (Fig. 1). Filtration was then used to prepare the product for loading onto the first column. The type of filter used (i.e. composition and pore size) and its effective product recovery was assayed by the quantitative anion exchange assay described in Example 3, Procedure A.
  • Centrifugation was followed by filtration through a Gelman Sciences Acrodisc™ 0.8/0.2 µm 2-stage syringe filter. Recovery of ACN53 depended on the pore size and type of membrane used for filtration. Filters such as polysulfone, PVDF membranes and cellulose acetate based membranes were used. Polysulfone and PVDF were preferred.
  • The supernatent from this step was made 2 mM in MgCl2, 2% (wt/vol) in sucrose and 2.5% (wt/vol) β-cyclodextrin. Benzonase™ (American International Chemical, Inc.) was added to a final concentration of 100 units/mL and allowed to incubate for 1 hr at room temperature. The treated material was clarified by centrifugation in a Beckman TJ-6 at 3000 rpm for 10 min and filtration through a Gelman Sciences Acrodisc™ 0.8/0.2 µm filter. The resultant supernatant was taken on to Example 2.
  • Example 2 Anion Exchange Chromatography
  • Overall, the characteristics of DEAE chromatography were found to be consistent, and loading studies with high titer lysate (3 ×1012 ACN53 particles/mL) showed a linear response between volume injected and ACN53 peak area recovered. Elution of a DEAE column by introduction of a linear salt gradient gave three major peaks. The first of these was a protein peak with a A260/A280 ratio of ca. 0.5. Next was the ACN53 peak (A260/A280 = 1.23) followed by a DNA (A260/A280 = 2) peak at the end of the gradient. The same three peaks were obtained whether run in a HEPES, Tris buffer or phosphate buffer system. The pH can be varied, however, if run at pH 7.5 using HEPES pH 7.5/NaCl/sucrose/MgCl2 less contaminating material bound to the columns. DEAE chromatography yielded a high degree of initial purification. The immobilized metal ion purification chromatography step removed these contaminants.
  • Column resins were tested for their separation characteristics in 6.6 x 50 mm (1.7 mL) borosilicate Omnifit™ columns (Omnifit Ltd., Cambridge, England). The columns were mounted on a PerSeptive Bioystems Biocad™ (Cambridge, MA) chromatography workstation. The chromatography was monitored on-line for pH, conductivity, and dual wavelength optical density detection at 280nm and 260nm.
  • Anion exchange resins were equilibrated in 50 mM HEPES, pH 7.5, 300 mM NaCl, 2 mM MgCl2, 2% sucrose at 1 mL/min. A 50 mM Tris buffer pH 8.0 (with 300 mM NaCl, 2 mM MgCl2, 2% sucrose) was also used. After the cell lysate was loaded and washed to baseline as monitored by absorbance, elution was performed with a 20 column volume 300-600 mM linear NaCl gradient and collected in 0.5 mL fractions. In preparation for future use, the column was then cleaned with 2 column volumes of 0.5 M NaOH, a 1.5 M NaCl wash and re-equilibrated.
  • In order to obtain an approximate control retention time, CsCl-ACN53 was injected onto a DEAE column equilibrated in 50mM Tris pH 8 at 2 mL/min (350 cm/hr) and eluted with a 10 min (11.7 column volume) 0-1.5 M linear NaCl gradient. A single peak was detected with an A260/A280 of 1.23. The protein bands present in this fraction reacted with Ad 5 polyclonal antibody upon slot-blot analysis.
  • Several peaks were resolved when an infected cell lysate sample was applied to the DEAE column (Fig. 2). The composition of the peaks were deduced from the A260/A280absorbance ratio. For example, the first peak has an A260/A280 ratio of 0.5, and is mainly protein. The third peak has an A260/A280 ratio of 2, suggesting that this material was nucleic acid. The ACN53 virus peak eluted second with a ratio of 1.23. The identities of these peaks were confirmed by spiking experiments and by running SDS gels of each peak. In typical experiments using the above conditions an A260/A280ratio of 1.23 ±.08 was found to be characteristic of virus peaks.
  • In order to assess the purification capabilities of DEAE chromatography, experiments were performed in which both non-infected ATCC-293 cell lysate and CsCI-ACN53 were applied to the column (Fig. 3). Most of the host cell material either passed through the column during the load or eluted at an earlier retention time than that of ACN53; however, a small peak eluted with the same retention time as ACN53. From these data it appeared that non-viral contamination of the ACN53 peak might be expected from host cell material. Examination of the contaminant peak, the peak eluting last in the cell lysate sample shown in Fig. 3, revealed an A260/A280 nm ratio of approximately 2. This indicated that the peak had a high nucleic acid content and could possibly be reduced or eliminated by treatment with nuclease. DEAE runs with and without Benzonase™ pretreatment demonstrated that the enzyme reduced the amount of contamination of host cell material in the ACN53 fraction pool.
  • Example 3 Immobilized Metal Affinity Chromatography
  • Immobilized metal affinity chromatography (IMAC) using zinc as the divalent cation (IZAC) gave higher product recovery and did not require any sample manipulation of the DEAE fraction pool prior to loading. Impurities removed by this method eluted in the flow through peak and were well resolved from product, leading to simpler pooling criteria. IZAC was reproducible, and when used with DEAE provided a two column purification protocol capable of delivering pure ACN53 as specified by SDS-PAGE gels and westerns, A260A280 ratios and infectious to non-infectious particle ratios. A summarized overall protocol is as follows:
    Harvest cells
    Nuclease treatment β-Clyclodextrin treatment
    Clarification
    DEAE chromatograhy
    IZAC chromatography
    Buffer exchange
    Stepwise recoveries in terms of total particles and infectious units are summarized in Tables 1 and 2 in the experimental section.
  • The Immobilized zinc affinity chromatography (IZAC) system was prepared for metal charging by washing the column sequentially with 1 volume of 100 mM EDTA and 1 volume of 0.2 M NaOH, flushing with water after each step. The matrix was subsequently charged with zinc by injecting 1 column volume of 100 mM ZnCl2 in H 20 acidified with 0.5 µL/mL glacial acetic acid. The matrix was then thoroughly washed in water prior to equilibration in 50 mM HEPES pH 7.5/450 mM NaCl/2% sucrose/2 mM MgCl2. Sample loading did not require any manipulation; DEAE pool fractions or CsCl derived material could be injected directly onto the column. After loading, the column was washed with a 10 column volume decreasing NaCl linear gradient from 50 mM HEPES pH 7.5/450 mM NaCl/2% sucrose/2 mM MgCl2 to 50 mM HEPES pH 7.5/150 mM NaCl/2% sucrose/2 mM MgCl2. Elution was performed with a linear 0-500 mM glycine gradient (in 150 mM NaCl) applied over 10 column volumes.
  • The interaction of ACN53 with a metal affinity column was shown to be metal specific (with zinc preferred); injection of CsCl-ACN53 onto an uncharged column (a column not pre-loaded with zinc) resulted in a shift of the product peak to the flow through. An ACN53-DEAE from Example 2 fraction pool purified over IZAC is shown in Fig.5. Analysis of an IZAC fraction pool produced a yield of 49-65% and an A260/A280 ratios of 1.22-1.25. A gel and western blot comparison of CsCl-ACN53, DEAE purified and DEAE/IZAC purified material can be seen in Figs 6 and 7. The CsCl-ACN53 and DEAE/IZAC materials were very similar, and the DEAE-only purified material was less pure by these criteria.
  • The effect of different IZAC buffer and elution systems was evaluated by splitting a DEAE-ACN53 pool in half and purifying both halves over IZAC in HEPES pH 7.5 and Tris pH 8 buffer systems. IZAC was also run in the presence of sucrose and MgCl2 without affecting the separation. The use of copper as the metal ion and imidazole as the elution agent were also tested (for a general review of metal affinity chromatography, see Belew et.al. 1987; Kato et.al, 1986). Various systems: zinc/glycine, zinc/imidazole, copper/glycine and copper/imidazole, all worked on the metal affinity purification columns. ACN53 can be eluted using a pH 7 to pH9 gradient.A: Assay for Recombinant Adenoviral Particle Number by Anion Exchange HPLC.
  • A 1 mL Resource Q- (Pharmacia, Piscataway, NJ) anion exchange column was used to quantitate the number of viral particles in various samples. The column was equilibrated in 300 mM NaCl, 50 mM HEPES, pH 7.5. at a flow rate of 1 mL /min on a Waters 625 chromatography system equipped with 717plus autosampler and a 991 photodiode array detector (PDA). The chromatography was monitored on the PDA detector (Milford, MA) scanning from 210 to 300 nm. A standard curve was constructed by injecting CsCl purified ACN53 virions characterized for total particles at a selected absorbance, A260 in 0.1%SDS.
  • The assay was independent of injected sample volume. After the sample loaded, the column was washed with two column volumes of equilibration buffer followed by a linear gradient from 300 to 600 mM NaCl in 50 mM HEPES, pH 7.5, over 10 column volumes. The gradient was followed with a 2 column volume wash with 600 mM NaCl in 50mM HEPES, pH 7.5. After each chromatographic run, the column was cleaned with 2.6 column volumes of 1.5 M NaCl in 50 mM HEPES, pH 7.5, and then re-equilibrated for the next injection. The column was cleaned more vigorously after injection of crude samples by injecting 0.25 to 1 column volume of 0.5 N NaOH followed by a wash with 1.5 M salt. Injecting NaOH and then running the gradient was a convenient way to accomplish cleaning. The use of such an assay in measuring the total number of adenovirus particles present are set forth in the following Table 1. Table 1. Yield and Purity Data Based on Total ACN53 Particles
    Step Viral Particle #/mL a Vol. (ml) Total Particles Step Yield b Total Yield b
    Lysate (Procedure 2) 6x1011 5.0 3x10 12 100%
    DEAE load (Example 1) 6x1011 5.0 3x1012
    DEAE eluate (Example 2) 4x1011 5.0 2x1012 67%
    IZAC load (Example 3) 4x1011 3.8 1.52x1012
    IZAC eluatec (Example 3) 2.38x1011 3.0 7.14x1011 47% 31%
    aAs determined by Analyzical Ion Exchange
    bIn terms of viral particle number
    cAfter dialysis into final formulation
  • B: Buffer Conditions
  • A variety of buffer conditions have been used in the purification of ACN53 by column chromatography in this study in the pH range of 7.0-8.0. (e.g. Seth Virology 68:1204-1206 (1994)). Checking for degradation at various pH limits is accomplished by assaying for degradation by TCID50 (Procedure C below) and analytical anion exchange analysis (Procedure A above). The effect of buffer salt concentration on the viability of ACN53 showed that abrupt changes of ionic strength can lead to loss of virus. Salt concentration should be carefully controlled and monitored.
  • C: Measurement of Infectious Recombinant Adenovirus Particles by TCID 50 Assay.
  • The quantitation of infectious Adenovirus Type 5 particles, before, during and after the purification methods taught in these Examples is accomplished by an end point titer assay (tissue culture infective dose-50%; abbreviated TCID50). (See Philipson, Virology 15:263-268 (1961)). Reagents, a materials list and instructions are available from Chemicon International, Inc. (cat.# 3130: "Adenovirus Direct Immunofluorescence Assay", Temecula, CA).
  • ATCC 293 cells were plated into a 96-well plate: 100 µL of 5 x 105 cells/mL for each well in complete MEM (10% bovine calf serum; 1% glutamine) media. In a separate plate, a 250 µL aliquot of virus sample diluted 1:106 was added to the first column and is serially diluted two-fold across the plate. One row was reserved for a positive control (CsCl-purified ACN53); one for a negative control. A 100 µL aliqout of each well was transferred to its identical position in the ATCC-293 seeded plate and allowed to incubate a 37° in a humidified incubator for 2 days. The media was then decanted by inversion and the cells were fixed with the addition of 50% acetone/50% methanol. After washing with PBS, the fixed cells were incubated for 45 min with a FITC labeled anti-ad5 antibody (Chemicon International #5016) prepared according to the kit instructions. After washing with PBS, the plate was examined under a fluorescent microscope (490 nm excitation, 520 nm emission) and scored for the presence of label. The titer was quantitated using the Titerprint Analysis program (Lynn, BioTechniques, 12:880-881 (1994)). Results from assays performed in accordance with this procedure are set forth in the following Table 2, which Table includes a ratio calculated using the values from Table 1 above.Table 2. Yield and Purity Data Based on Infective ACN53 Particles
    Step Step Yield (TCID 50) A 260 :A 280 Ratio - Viral Particle Purity by HPLC at A 280
    Lysate - - 3%
    DEAE load 49% 1.17 92%
    IZAC Eluate 44% 1.23 98%
  • The ratio of total virus particles to infectious viral particles can vary widely from preparation to preparation. Values for CsCl derived viruses in the range of 60 to 80. The calculation of this ratio in crude lysates or semi-pure fractions has been made possible by the anion exchange particle assay (see Example 3[A]). Using analytical anion exchange changes in the ratio of ideal virus particles to infectious virus particles can be monitored. Using the purification method of this invention the total virus particle to infectious virus particle ratio of the crude lysate and subsequently purified material is comparable to that obtained using ultracentrafugation. Within the error of the assay these values are equivalent. By this criteria, chromatographic purification is well tolerated by the virus. By other criteria, namely SDS-PAGE and western blot analysis (Figs. 6 and 7), chromatographic purification is superior to ultracentrifugation based methods.
  • D: Assay for Infectious Recombinant Adenovirus Particles by Expression of P53 Protein.
  • The activity of virus preparations was also tested by assaying for the expression of the p53 gene product in Saos-2 cells, a p53-negative osteosarcoma cell line. Saos-2 cells were seeded into a 6-well tissue culture plate at a concentration of 5 x 105 cells/well in 3 mL of media: Kaighn's nutrient mixture F12: DME High Glucose (1:1 mixture), supplemented with 2 mM L-glutamine and 10% fetal bovine serum. The cells were incubated in humidified air, 7±1% CO2 at 37° for 16-24 hrs. The spent media were removed and replaced with 1 mL of fresh media, and the cells were infected at 20,40, or 60 MOI of purified virus. After incubation for 1 hr, an additional 2 mL of media is added and allowed to incubate for 8 hrs. The cells were then washed once with Dulbecco's-PBS and lysed by adding of 250 µL of: 50 mM tris/0.5% Noridet P-40/250 mM NaCl/5 mM EDTA/5 mM NaF/5 µg/mL Leupeptin and 5 µg/mL Aprotinin/2 mM PMSF. The plate was incubated on ice for 5 min after which the lysates were transferred to individual 1.7 mL microcentrifuge tubes. They were spun down for 45 sec at 14000 rpm in a microfuge and the supernatants were assayed for the presence of p53 protein by western blot analysis with the primary anti-p53 monoclonal antibody 1801 (Vector Laboratories, Burlingame, California) and a 1:1 mixture of sheep anti-mouse IgG-HRP and streptavidin-HRP (Amersham). The p53 protein band was detected using Amersham's ECL detection kit according to the manufacturer's instructions. The results of the use of such assay are shown in Figure 8. p53 expression can be seen in ACN53 purified only by DEAE chromatography as well as by both DEAE and IZAC chromatography. (The lower levels of expression in lanes 5 and 6 are due to the fact that these samples were run at a lower MOI than the DEAE samples.)
  • E: Host Cell Protein Contamination
  • Host cell contamination was assessed by western blot analysis using polyclonal antibodies developed against ATCC-293 cell components. Polyclonal antibodies were commercially obtained and were raised against various 293 cellular antigens (HTI, Ramona, CA). The results indicated that the final products of either CsCl or DEAE-IZAC purification contained no detectable host cell contaminants. In the case of semi-pure DEAE-ACN53, there was a small amount of immunogenic contamination seen in the product pool: 3 major bands and several minor ones. The majority of host cell contamination is recovered in the flow-through portion of the DEAE step. Contaminants which copurified with ACN53 in the DEAE step were removed by zinc affinity chromatography, and were recovered in IZAC flow-through fractions prior to the introduction of the glycine gradient.
  • F: SDS-PAGE Analysis Samples Containing Recombinant Adenovirus Particles.
  • For Coomassie blue staining, 100-200 µL of an adenovirus containing sample (at approximately 1x1011 particles/mL) was collected, desalted by trichloroacetic acid precipitation or by dialysis followed by concentration in a speed-vac. The sample was then resuspended in SDS-PAGE reducing buffer (125 mM Tris-HCL, pH 6.8, 20% glycerol, 4% (w/v) SDS, 0.005% bromophenol blue, 0.5% β- mercaptoethanol) to approximately 30 µL, boiled for 5 min and loaded onto a 1 mm x 10 well Novex 8-16% gradient Tris-Glycine minigel. Samples were electrophoresed for 1.5 hrs at 140 V. The gel is then fixed in 40% methanol/10% acetic acid for 30 min, and then Coomassie stained with the Pro-Blue staining system (Integrated Separation Systems, Natick MA.) according to the vendor's procedure.
  • Gels to be silver stained were loaded with 5-15 µL of sample. The sample was boiled with an equal volume of reducing buffer and electrophoresed as described for Coomassie detection. Fixing was performed by treating the gel in 10% trichloroacetic acid for 1 hr followed by a 3x wash in water purified to 18 megohms. The gel was stained with the Daiichi silver staining kit according to the instructions provided (Integrated Separation Systems).
  • Example IV Vector Infectivity. Transfer of Therapeutic Gene
  • Characterization of DEAE-IZAC-ACN53 has shown that virions retained their infectivity during chromatographic treatment as measured by TCID50 analysis (Table 1,2) and were able to effect gene transfer as assayed by P53 protein expression in Saos-2 cells (Fig 8). Comparison of this DEAE-IZAC-ACN53 to CsCl-ACN53 in SDS-PAGE analysis has shown that there were more lower molecular weight protein bands present in the CsCl-ACN53 (Figs. 6 and 7). Side-by-side comparison of different CsCl-ACN53 lots has shown some batch-to-batch variability whereas chromatographically produced material has been very consistent. The total to infectious particle ratio and absorbance spectrum of both types of material are directly comparable. Characterization of chromatographically produced ACN53 in terms of purity and activity have shown that a 1 day 2-column procedure can replace a 3 day ultracentrifugation protocol.
  • A preferred purification scheme for ACN53 as shown above is to treat infected cell lysate with Benzonase™ prior to the chromatographic steps. Clarification is then accomplished by step filtration through 0.8 µm followed by 0.2 µm membranes. If necessary, a larger pore (e.g. 5 µm) prefiltration step can be added for more viscous suspensions. Adjustment to pH 7.5/300 mM NaCl is then performed in preparation for loading onto a DEAE column. The product peak, as detected by the A260/A280 nm ratio or the characteristic photo-diode array spectrum, is pooled and directly injected onto a zinc-charged, iso-osmotically equilibrated metal affinity column. The ionic strength of the buffer is then gradually lowered to approximate phosphate-buffered saline (ca,150 mM NaCl) prior to elution of product with a glycine gradient. This material is then dialyzed into the final formulation.

Claims (15)

  1. A method for purification of a recombinant viral vector containing a therapeutic gene from a cell lysate, which comprises:
    a) treating said cell lysate with an enzymatic agent that selectively degrades both unencapsulated DNA and RNA;
    b) chromatographing the treated lysate from step a) on a first resin; and
    c) chromatographing the eluant from step b) on a second resin;
    wherein one resin is an anion exchange resin and the other is an immobilized metal ion affinity resin.
  2. A method of claim 1, comprising the additional step of filtering the treated lysate from step (a).
  3. A method of claim 1 or claim 2 comprising the additional step of buffering the pH of the cell lysate between 5.0 and 9.0 before applying it to the first resin.
  4. A method of any one of claims 1 to 3 wherein the first resin is an anion exchange resin and the second resin is an immobilized metal affinity resin.
  5. A method of any one of claims 1 to 3, wherein the recombinant viral vector is a retrovirus or an adenovirus.
  6. A method of claim 5, wherein the therapeutic gene contained in the recombinant adenoviral vector is a tumor suppressor gene.
  7. A method of claim 6, wherein the tumor suppressor gene is wild-type p53.
  8. A method of claim 5 or claim 6, wherein the recombinant adenoviral vector is a type 2 or type 5 adenovirus.
  9. A method of claim 4, wherein the anion exchange resin is chosen from DEAE, TMAE, DMAE, QAE and PEI and/or wherein the immobilized metal affinity resin is charged with a divalent cation of a metal chosen from cobalt, nickel, copper, zinc, calcium and magnesium.
  10. A method of claim 9. wherein the immobilized metal affinity resin is a TED or an IDA resin.
  11. A method of claim 10 wherein the IDA resin is an IDA- (cross-linked agarose) resin.
  12. A method of any one of claims 1 to 11, wherein the enzymatic agent that selectively degrades both unencapsulated DNA and RNA is one or more enzymes.
  13. A method of claim 12, wherein the enzymatic agent is an endonuclease.
  14. A method of claim 13, wherein the endonuclease is Benzonase.
  15. A method for purification of recombinant viral vectors containing a therapeutic gene from a cell lysate, which comprises:
    a) treating said cell lysate with an enzymatic agent that selectively degrades both unencapsulated DNA and RNA;
    b) chromatographing the treated lysate from step a) on a first resin; and
    d) chromatographing the eluant from step b) on a second resin;
    wherein:
    one resin is an anion exchange resin and the other is a hydrophobic interaction chromatography resin.
EP96911282A 1995-03-07 1996-03-06 Method of purification of recombinant viral vectors containing a therapeutic gene Expired - Lifetime EP0813606B1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
EP06075526A EP1686180A3 (en) 1995-03-07 1996-03-06 Method of purification of recombinant viral vectors containing a therapeutic gene

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US08/400,793 US5837520A (en) 1995-03-07 1995-03-07 Method of purification of viral vectors
US400793 1995-03-07
PCT/US1996/003369 WO1996027677A2 (en) 1995-03-07 1996-03-06 Method of purification of recombinant viral vectors containing a therapeutic gene

Related Child Applications (1)

Application Number Title Priority Date Filing Date
EP06075526A Division EP1686180A3 (en) 1995-03-07 1996-03-06 Method of purification of recombinant viral vectors containing a therapeutic gene

Publications (2)

Publication Number Publication Date
EP0813606A2 EP0813606A2 (en) 1997-12-29
EP0813606B1 true EP0813606B1 (en) 2006-06-07

Family

ID=23585038

Family Applications (2)

Application Number Title Priority Date Filing Date
EP06075526A Withdrawn EP1686180A3 (en) 1995-03-07 1996-03-06 Method of purification of recombinant viral vectors containing a therapeutic gene
EP96911282A Expired - Lifetime EP0813606B1 (en) 1995-03-07 1996-03-06 Method of purification of recombinant viral vectors containing a therapeutic gene

Family Applications Before (1)

Application Number Title Priority Date Filing Date
EP06075526A Withdrawn EP1686180A3 (en) 1995-03-07 1996-03-06 Method of purification of recombinant viral vectors containing a therapeutic gene

Country Status (12)

Country Link
US (1) US5837520A (en)
EP (2) EP1686180A3 (en)
JP (2) JP3858200B2 (en)
AR (1) AR001177A1 (en)
AT (1) ATE329045T1 (en)
AU (1) AU5421396A (en)
DE (1) DE69636217T2 (en)
DK (1) DK0813606T3 (en)
ES (1) ES2264801T3 (en)
PT (1) PT813606E (en)
WO (1) WO1996027677A2 (en)
ZA (1) ZA961849B (en)

Families Citing this family (126)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH09508017A (en) * 1994-01-12 1997-08-19 ジェネティック セラピー,インコーポレイテッド Purification of retrovirus vector
FR2737730B1 (en) * 1995-08-10 1997-09-05 Pasteur Merieux Serums Vacc PROCESS FOR PURIFYING VIRUSES BY CHROMATOGRAPHY
CA2625279A1 (en) * 1995-08-30 1997-03-06 Genzyme Corporation Chromatographic purification of adenovirus and aav
US20040014709A1 (en) * 1996-01-08 2004-01-22 Canji, Inc. Methods and compositions for interferon therapy
US6392069B2 (en) * 1996-01-08 2002-05-21 Canji, Inc. Compositions for enhancing delivery of nucleic acids to cells
US6485958B2 (en) * 1996-07-01 2002-11-26 Gencell Sa Method for producing recombinant adenovirus
US6576133B2 (en) 1996-11-13 2003-06-10 Transgenomic, Inc Method and system for RNA analysis by matched ion polynucleotide chromatography
US6475388B1 (en) 1996-11-13 2002-11-05 Transgenomic, Inc. Method and system for RNA analysis by matched ion polynucleotide chromatography
EP0968284B1 (en) * 1996-11-20 2006-12-13 Introgen Therapeutics, Inc. An improved method for the production and purification of adenoviral vectors
US7732129B1 (en) * 1998-12-01 2010-06-08 Crucell Holland B.V. Method for the production and purification of adenoviral vectors
US20080261289A1 (en) * 1996-12-13 2008-10-23 Schering-Plough Corporation Compositions comprising viruses and methods for concentrating virus preparations
US6261823B1 (en) 1996-12-13 2001-07-17 Schering Corporation Methods for purifying viruses
US6544769B1 (en) * 1996-12-13 2003-04-08 Schering Corporation Compostions comprising viruses and methods for concentrating virus preparations
US6177559B1 (en) 1998-04-24 2001-01-23 Transgenomic, Inc. Process for separation of polynucleotide fragments
US6566118B1 (en) 1997-09-05 2003-05-20 Targeted Genetics Corporation Methods for generating high titer helper-free preparations of released recombinant AAV vectors
PT1944362E (en) 1997-09-05 2016-01-27 Genzyme Corp Methods for generating high titer helper-free preparations of recombinant aav vectors
US6995006B2 (en) 1997-09-05 2006-02-07 Targeted Genetics Corporation Methods for generating high titer helper-free preparations of released recombinant AAV vectors
AU1186399A (en) 1997-10-09 1999-05-03 Biogen, Inc. Chromatographically concentrated baculovirus and methods
US6258264B1 (en) 1998-04-10 2001-07-10 Transgenomic, Inc. Non-polar media for polynucleotide separations
US6265168B1 (en) 1998-10-06 2001-07-24 Transgenomic, Inc. Apparatus and method for separating and purifying polynucleotides
MXPA00010338A (en) * 1998-04-22 2005-06-17 Genvec Inc Efficient purification of adenovirus.
US6489305B1 (en) * 1998-05-08 2002-12-03 Canji, Inc. Methods and compositions for the treatment of ocular diseases
DK1930418T3 (en) 1998-09-04 2015-07-13 Genzyme Corp Methods for Generating Helper-Free High Titer Preparations of Released Recombinant AAV Vectors
US7691370B2 (en) * 1998-10-15 2010-04-06 Canji, Inc. Selectivity replicating viral vector
US6689600B1 (en) * 1998-11-16 2004-02-10 Introgen Therapeutics, Inc. Formulation of adenovirus for gene therapy
AU779267B2 (en) 1998-12-31 2005-01-13 Centelion S.A.S. Method for separating viral particles
EP1155120B1 (en) * 1999-02-22 2006-07-05 Transgene S.A. Method for obtaining a purified viral preparation
DE19933719A1 (en) * 1999-07-19 2001-01-25 Medigene Ag Structural protein in adeno-associated virus with altered chromatographic properties, its production and use
US6464976B1 (en) 1999-09-07 2002-10-15 Canji, Inc. Methods and compositions for reducing immune response
US6316185B1 (en) 1999-09-29 2001-11-13 Mountain View Pharmaceuticals, Inc. Quantitation of viruses by light scattering
JP4684434B2 (en) * 2000-02-16 2011-05-18 Jsr株式会社 Virus concentration particle, virus concentration reagent, virus concentration method and virus detection method
DE10010342A1 (en) 2000-03-06 2001-09-20 Merck Patent Gmbh Method for reducing the endotoxin content of nucleic acid (I) is derived from natural, genetic engineering or biotechnological sources is used to produce high-purity plasmid DNA from natural sources
CA2410948C (en) * 2000-05-31 2012-07-17 Chiron Corporation Method for the purification of alphavirus replicon particles
US6593123B1 (en) 2000-08-07 2003-07-15 Avigen, Inc. Large-scale recombinant adeno-associated virus (rAAV) production and purification
US6447995B1 (en) 2000-10-04 2002-09-10 Genvec, Inc. Utilizing intrinsic fluorescence to detect adenovirus
US20020064860A1 (en) * 2000-11-29 2002-05-30 Schering Corporation Method for purifying adenoviruses
WO2002049422A2 (en) * 2000-12-20 2002-06-27 K.U. Leuven Research And Development Non-human animal disease models
KR100451308B1 (en) * 2001-12-28 2004-10-06 선바이오(주) A virus removal process in the purification of biologically active molecules
US6833238B2 (en) 2002-01-04 2004-12-21 Applera Corporation Petal-array support for use with microplates
CA2476447A1 (en) * 2002-02-20 2003-08-28 Merck & Co., Inc. Method of determining adenovirus particle concentration
WO2003085138A1 (en) * 2002-03-29 2003-10-16 Merck & Co., Inc. Methods of virus production
EP1492890A4 (en) * 2002-03-29 2006-10-18 Merck & Co Inc Large scale methods of producing adenovirus and adenovirus seed stocks
AU2003229060A1 (en) * 2002-05-14 2003-12-02 Merck And Co., Inc. Methods of adenovirus purification
US20030224354A1 (en) * 2002-05-30 2003-12-04 Introgen Therapeutics Inc. Quantifying viral particles with intrinsic fluorescence
US20040018559A1 (en) * 2002-07-26 2004-01-29 Applera Corporation Size-exclusion ion-exchange particles
US20040106184A1 (en) * 2002-08-28 2004-06-03 Introgen Therapeutics Inc. Chromatographic methods for adenovirus purification
CA2529053A1 (en) * 2003-06-18 2004-12-29 Onyx Pharmaceuticals, Inc. Method for purifying virus
ES2359473T3 (en) 2003-07-21 2011-05-23 Transgene S.A. MULTIFUNCTIONAL CYTOKINS.
KR101167330B1 (en) * 2003-11-24 2012-07-23 캔지, 인크. A pharmaceutical composition for reducing dermal scarring but not for inhibiting wound closure
CA2551026A1 (en) * 2003-12-23 2005-07-14 Schering Corporation Methods for producing a549 cell lines stable in serum-free medium suspension culture
US7479222B2 (en) * 2004-02-05 2009-01-20 Millipore Corporation Porous adsorptive or chromatographic media
US20060160122A1 (en) * 2004-02-18 2006-07-20 Applera Corporation Polyelectrolyte-coated size-exclusion ion-exchange particles
US20050196856A1 (en) * 2004-02-18 2005-09-08 Applera Corporation Polyelectrolyte-coated size-exclusion ion-exchange particles
US20050181378A1 (en) * 2004-02-18 2005-08-18 Applera Corporation Polyelectrolyte-coated size-exclusion ion-exchange particles
DK1780269T3 (en) * 2004-02-23 2009-10-12 Crucell Holland Bv Virus Purification Methods
WO2006011580A1 (en) * 2004-07-27 2006-02-02 Genomidea, Inc. Method of purifying virus envelope
WO2006052302A2 (en) * 2004-11-03 2006-05-18 Introgen Therapeutics Inc. Method of producing and purifying adenoviral vectors
WO2006065827A2 (en) * 2004-12-13 2006-06-22 Canji, Inc. Cell lines for production of replication-defective adenovirus
DE602006003420D1 (en) * 2005-04-11 2008-12-11 Crucell Holland Bv VIRUS CLEANING WITH ULTRA FILTRATION
EP1736538A1 (en) * 2005-06-21 2006-12-27 Cytos Biotechnology AG Process for the preparative purification of virus-like-particles (VLPs)
EP1907537A4 (en) * 2005-07-14 2010-11-10 Mayo Foundation Paramyxoviridae virus preparations
DE602006019916D1 (en) * 2005-12-12 2011-03-10 Canji Inc ADENOVIRAL EXPRESSION VECTORS WITH AN EXPRESSION CASSETTE IN THE E1 AREA AND AN INACTIVATED E2B POLYMERASE
WO2008060356A2 (en) * 2006-09-29 2008-05-22 Canji, Inc. Methods and compositions for gene therapy
EP2390340A3 (en) 2007-01-30 2012-02-22 Transgene SA vector encoding Papillomavirus E1 and E2 polypeptides with reduced percentage of identity
US9433922B2 (en) 2007-08-14 2016-09-06 Emd Millipore Corporation Media for membrane ion exchange chromatography based on polymeric primary amines, sorption device containing that media, and chromatography scheme and purification method using the same
US20090130738A1 (en) * 2007-11-19 2009-05-21 Mikhail Kozlov Media for membrane ion exchange chromatography
PL2307551T3 (en) 2008-06-18 2017-07-31 Oxford Biomedica (Uk) Limited Purification of retroviral vectors
DK2349520T3 (en) * 2008-10-27 2016-08-15 Glaxosmithkline Biologicals Sa Purification Procedure for Group A Streptococcus Carbohydrate
KR101504392B1 (en) * 2008-11-03 2015-03-19 크루셀 홀란드 비.브이. Method for the production of adenoviral vectors
KR20120052352A (en) 2009-08-07 2012-05-23 트랜스진 에스.에이. Composition for treating hbv infection
AU2011214262B2 (en) 2010-02-15 2015-05-21 Crucell Holland B.V. Method for the production of Ad26 adenoviral vectors
CN102985536B (en) 2010-04-14 2017-12-05 Emd密理博公司 Produce high-titer, high-purity virus stocks method and use its method
WO2012038367A1 (en) 2010-09-20 2012-03-29 Crucell Holland B.V. Therapeutic vaccination against active tuberculosis
BR112013004582A2 (en) 2010-09-27 2016-09-06 Crucell Holland Bv method for inducing an immune response in a subject against a parasite antigen that causes malaria
DE102010046817A1 (en) 2010-09-28 2012-03-29 Sartorius Stedim Biotech Gmbh A method for separating viruses from a contaminant-containing liquid medium
EP3202897B1 (en) 2010-12-09 2020-04-15 Institut Pasteur Diagnostic use of a fusion polypeptide comprising a viral protein and a mgmt enzyme
US9610354B2 (en) 2011-04-18 2017-04-04 National Center Of Neurology And Psychiatry Drug delivery particle and method for producing the same
TWI623618B (en) 2011-07-12 2018-05-11 傳斯堅公司 Hbv polymerase mutants
WO2013045658A1 (en) 2011-09-29 2013-04-04 Transgene Sa Immunotherapy composition and regimen for treating hepatitis c virus infection
TW201321016A (en) 2011-09-29 2013-06-01 Transgene Sa Immunotherapy composition and regimen for treating hepatitis C virus infection
EP2764094A1 (en) * 2011-10-05 2014-08-13 MolMed SpA Viral vectors purification system
US20130122038A1 (en) 2011-11-14 2013-05-16 The United States Of America As Represented By The Secretary Of The Department Heterologous prime-boost immunization using measles virus-based vaccines
DK2825640T3 (en) 2012-03-12 2016-08-01 Crucell Holland Bv BATCHES OF RECOMBINANT ADENOVIRUS WITH CHANGED TERMINAL END
US8932607B2 (en) 2012-03-12 2015-01-13 Crucell Holland B.V. Batches of recombinant adenovirus with altered terminal ends
US9125870B2 (en) 2012-03-22 2015-09-08 Crucell Holland B.V. Vaccine against RSV
AP2014007993A0 (en) 2012-03-22 2014-10-31 Crucell Holland Bv Vaccine against RSV
CN103571800B (en) * 2012-07-27 2018-04-24 江苏康润生物科技有限公司 A kind of method of host DNA in removal vaccine
EP2912069B1 (en) 2012-10-23 2019-07-31 Emory University Gm-csf and il-4 conjugates, compositions, and methods related thereto
CN105188745B (en) 2013-04-25 2019-10-18 扬森疫苗与预防公司 RSV F polypeptide before stabilized soluble fusion
EP3010931B1 (en) 2013-06-17 2018-06-13 Janssen Vaccines & Prevention B.V. Stabilized soluble pre-fusion rsv f polypeptides
WO2016131945A1 (en) 2015-02-20 2016-08-25 Transgene Sa Combination product with autophagy modulator
AU2016249798B2 (en) 2015-04-14 2022-05-26 Janssen Vaccines And Prevention B.V. Recombinant adenovirus expressing two transgenes with a bidirectional promoter
PL3319633T3 (en) 2015-07-07 2021-04-19 Janssen Vaccines & Prevention B.V. Vaccine against rsv
US10457708B2 (en) 2015-07-07 2019-10-29 Janssen Vaccines & Prevention B.V. Stabilized soluble pre-fusion RSV F polypeptides
US9663766B2 (en) * 2015-07-24 2017-05-30 Bio-Rad Laboratories, Inc. Methods for purifying adenovirus vectors
WO2017174564A1 (en) 2016-04-05 2017-10-12 Janssen Vaccines & Prevention B.V. Vaccine against rsv
AU2017248021B2 (en) 2016-04-05 2021-08-12 Janssen Vaccines & Prevention B.V. Stabilized soluble pre-fusion RSV F proteins
CA3023022A1 (en) 2016-05-04 2017-11-09 Transgene Sa Combination therapy with cpg tlr9 ligand
AU2017264562B2 (en) 2016-05-12 2023-03-09 Janssen Vaccines & Prevention B.V. Potent and balanced bidirectional promoter
JP2019523644A (en) 2016-05-30 2019-08-29 ヤンセン ファッシンズ アンド プリベンション ベーフェーJanssen Vaccines & Prevention B.V. Stabilized pre-fusion RSV F protein
EP3472327B1 (en) 2016-06-20 2020-08-19 Janssen Vaccines & Prevention B.V. Potent and balanced bidirectional promoter
JP7229151B2 (en) 2016-07-14 2023-02-27 ヤンセン ファッシンズ アンド プリベンション ベーフェー HPV vaccine
JP7016136B2 (en) 2016-09-05 2022-02-04 国立研究開発法人科学技術振興機構 Methods and kits for detecting pathogenic microorganisms
US20190328869A1 (en) 2016-10-10 2019-10-31 Transgene Sa Immunotherapeutic product and mdsc modulator combination therapy
EP3541420A2 (en) 2016-11-16 2019-09-25 Immunomic Therapeutics, Inc. Nucleic acids for treatment of allergies
JP6721797B2 (en) 2017-02-09 2020-07-15 ヤンセン ファッシンズ アンド プリベンション ベーフェーJanssen Vaccines & Prevention B.V. Strong and short promoter for heterologous gene expression
KR102637960B1 (en) 2017-04-22 2024-02-21 이뮤노믹 쎄라퓨틱스, 인크. Improved LAMP construct
EP3618854A1 (en) 2017-05-02 2020-03-11 Immunomic Therapeutics, Inc. Lamp (lysosomal associated membrane protein) constructs comprising cancer antigens
CA3061278A1 (en) 2017-05-17 2018-11-22 Janssen Vaccines & Prevention B.V. Methods and compositions for inducing protective immunity against rsv infection
WO2019053109A1 (en) 2017-09-15 2019-03-21 Janssen Vaccines & Prevention B.V. Method for the safe induction of immunity against rsv
US11603527B2 (en) 2017-12-27 2023-03-14 Global Life Sciences Solutions Usa Llc Method and kit for viral vector isolation
WO2019168200A1 (en) * 2018-03-02 2019-09-06 国立研究開発法人科学技術振興機構 Method for detecting enzymatic reaction product
GB201806736D0 (en) * 2018-04-25 2018-06-06 Ge Healthcare Bioprocess R&D Ab Method for virus purification
US20210261647A1 (en) 2018-05-15 2021-08-26 Immunomic Therapeutics, Inc Lamp constructs comprising allergens
JP7233880B2 (en) * 2018-10-22 2023-03-07 キヤノンメディカルシステムズ株式会社 Viral RNA isolation method and virus quantification method
TW202043256A (en) 2019-01-10 2020-12-01 美商健生生物科技公司 Prostate neoantigens and their uses
EP3927833A4 (en) 2019-02-21 2022-11-30 Unleash Immuno Oncolytics, Inc. Oncolytic adenoviral vector and methods of use
AU2020366515A1 (en) 2019-10-18 2022-04-21 Immunomic Therapeutics, Inc. Improved lamp constructs comprising cancer antigens
AU2020385683A1 (en) 2019-11-18 2022-06-30 Janssen Biotech, Inc. Vaccines based on mutant CALR and JAK2 and their uses
TW202144389A (en) 2020-02-14 2021-12-01 美商健生生物科技公司 Neoantigens expressed in multiple myeloma and their uses
TW202144388A (en) 2020-02-14 2021-12-01 美商健生生物科技公司 Neoantigens expressed in ovarian cancer and their uses
WO2021209897A1 (en) 2020-04-13 2021-10-21 Janssen Biotech, Inc. Psma and steap1 vaccines and their uses
WO2022009049A1 (en) 2020-07-06 2022-01-13 Janssen Biotech, Inc. Prostate neoantigens and their uses
EP4176087A1 (en) 2020-07-06 2023-05-10 Janssen Biotech, Inc. A method for determining responsiveness to prostate cancer treatment
WO2022009052A2 (en) 2020-07-06 2022-01-13 Janssen Biotech, Inc. Prostate neoantigens and their uses
WO2023201201A1 (en) 2022-04-10 2023-10-19 Immunomic Therapeutics, Inc. Bicistronic lamp constructs comprising immune response enhancing genes and methods of use thereof

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4569794A (en) * 1984-12-05 1986-02-11 Eli Lilly And Company Process for purifying proteins and compounds useful in such process

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1984004696A1 (en) * 1983-05-31 1984-12-06 Baxter Travenol Lab Particle adsorption
US5447859A (en) * 1993-07-16 1995-09-05 Viagene Method for the purification or removal of retroviruses using sulfated cellulose

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4569794A (en) * 1984-12-05 1986-02-11 Eli Lilly And Company Process for purifying proteins and compounds useful in such process

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
ALBERTS ET AL: "Molecular Biology of the cell. 3rd ED.", 1994, GARLAND PUBLISHING, NEW YORK *
KLEMPERER H.G.: "Study of adenovirus antigens fractionated by chromatography on DEAE-cellulose", VIROLOGY, vol. 9, pages 536 - 545 *
MARAMOROSCH ET AL: "Methods in virology", 1967, ACADEMIC PRESS, NEW YORK *

Also Published As

Publication number Publication date
DK0813606T3 (en) 2006-10-02
EP1686180A3 (en) 2006-08-09
DE69636217T2 (en) 2007-04-26
JP2006271396A (en) 2006-10-12
US5837520A (en) 1998-11-17
AU5421396A (en) 1996-09-23
JP4182445B2 (en) 2008-11-19
AR001177A1 (en) 1997-09-24
ATE329045T1 (en) 2006-06-15
EP0813606A2 (en) 1997-12-29
PT813606E (en) 2006-10-31
EP1686180A2 (en) 2006-08-02
JP3858200B2 (en) 2006-12-13
JP2000510682A (en) 2000-08-22
DE69636217D1 (en) 2006-07-20
ES2264801T3 (en) 2007-01-16
WO1996027677A3 (en) 1997-01-16
WO1996027677A2 (en) 1996-09-12
ZA961849B (en) 1997-08-27

Similar Documents

Publication Publication Date Title
EP0813606B1 (en) Method of purification of recombinant viral vectors containing a therapeutic gene
JP5898261B2 (en) Purification method of pox virus
KR100762033B1 (en) Method for separating viral particles
AU2004249199B2 (en) Method for purifying virus
Graessmann et al. Expression of Epstein-Barr virus genes in different cell types after microinjection of viral DNA.
US7393631B2 (en) Method for purifying adenoviruses
EP1226264A2 (en) Cell lines and constructs useful in production of e1-deleted adenoviruses in absence of replication competent adenovirus
Stillman et al. An adenovirus protein associated with the ends of replicating DNA molecules
CA2214837C (en) Method of purification of recombinant viral vectors containing a therapeutic gene
CA2522630C (en) Method of purification of recombinant viral vectors containing a therapeutic gene
Hermiston et al. Construction of mutations in the adenovirus early region 3 (E3) transcription units
US6974694B2 (en) Adenoviruses for control of gene expression
Quinlan THE EXPRESSION AND LOCALIZATION OF THE MAJOR DNA-BINDING PROTEIN OF HSV (VIROLOGY, CELL BIOLOGY, NUCLEAR ANTIGEN, HERPES SIMPLEX VIRUS)
Jin et al. Expression of Human Papillomavirus Type 16 L1 in Baculovirus Expression Systems
Affinity Tagging Retrovirus Vectors with a Metal
FR2788064A1 (en) Separation of viral particles, useful e.g. for purification, titration or serotype identification, by chromatography on support having ion-exchange groups attached via flexible linker

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 19971007

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE CH DE DK ES FI FR GB GR IE IT LI LU MC NL PT SE

17Q First examination report despatched

Effective date: 20030626

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AT BE CH DE DK ES FI FR GB GR IE IT LI LU MC NL PT SE

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT;WARNING: LAPSES OF ITALIAN PATENTS WITH EFFECTIVE DATE BEFORE 2007 MAY HAVE OCCURRED AT ANY TIME BEFORE 2007. THE CORRECT EFFECTIVE DATE MAY BE DIFFERENT FROM THE ONE RECORDED.

Effective date: 20060607

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REF Corresponds to:

Ref document number: 69636217

Country of ref document: DE

Date of ref document: 20060720

Kind code of ref document: P

REG Reference to a national code

Ref country code: CH

Ref legal event code: NV

Representative=s name: KATZAROV S.A.

REG Reference to a national code

Ref country code: SE

Ref legal event code: TRGR

REG Reference to a national code

Ref country code: DK

Ref legal event code: T3

REG Reference to a national code

Ref country code: GR

Ref legal event code: EP

Ref document number: 20060402881

Country of ref document: GR

REG Reference to a national code

Ref country code: PT

Ref legal event code: SC4A

Effective date: 20060901

REG Reference to a national code

Ref country code: ES

Ref legal event code: FG2A

Ref document number: 2264801

Country of ref document: ES

Kind code of ref document: T3

ET Fr: translation filed
PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

26N No opposition filed

Effective date: 20070308

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: IT

Payment date: 20120323

Year of fee payment: 17

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: DK

Payment date: 20130226

Year of fee payment: 18

Ref country code: MC

Payment date: 20130227

Year of fee payment: 18

Ref country code: IE

Payment date: 20130306

Year of fee payment: 18

Ref country code: LU

Payment date: 20130318

Year of fee payment: 18

Ref country code: SE

Payment date: 20130308

Year of fee payment: 18

Ref country code: ES

Payment date: 20130313

Year of fee payment: 18

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: GR

Payment date: 20130227

Year of fee payment: 18

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: AT

Payment date: 20130225

Year of fee payment: 18

Ref country code: PT

Payment date: 20130227

Year of fee payment: 18

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: BE

Payment date: 20130410

Year of fee payment: 18

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: CH

Payment date: 20140127

Year of fee payment: 19

Ref country code: FI

Payment date: 20140305

Year of fee payment: 19

Ref country code: DE

Payment date: 20140331

Year of fee payment: 19

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: FR

Payment date: 20140225

Year of fee payment: 19

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: GB

Payment date: 20140225

Year of fee payment: 19

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: NL

Payment date: 20140311

Year of fee payment: 19

REG Reference to a national code

Ref country code: PT

Ref legal event code: MM4A

Free format text: LAPSE DUE TO NON-PAYMENT OF FEES

Effective date: 20140908

REG Reference to a national code

Ref country code: DK

Ref legal event code: EBP

Effective date: 20140331

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20140306

REG Reference to a national code

Ref country code: SE

Ref legal event code: EUG

REG Reference to a national code

Ref country code: AT

Ref legal event code: MM01

Ref document number: 329045

Country of ref document: AT

Kind code of ref document: T

Effective date: 20140306

REG Reference to a national code

Ref country code: GR

Ref legal event code: ML

Ref document number: 20060402881

Country of ref document: GR

Effective date: 20141002

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20140307

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: PT

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20140908

REG Reference to a national code

Ref country code: IE

Ref legal event code: MM4A

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: GR

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20141002

Ref country code: IE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20140306

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: AT

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20140306

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IT

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20140306

REG Reference to a national code

Ref country code: ES

Ref legal event code: FD2A

Effective date: 20150424

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: DK

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20140331

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: ES

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20140307

REG Reference to a national code

Ref country code: DE

Ref legal event code: R119

Ref document number: 69636217

Country of ref document: DE

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: FI

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20150306

REG Reference to a national code

Ref country code: CH

Ref legal event code: PL

GBPC Gb: european patent ceased through non-payment of renewal fee

Effective date: 20150306

REG Reference to a national code

Ref country code: NL

Ref legal event code: MM

Effective date: 20150401

REG Reference to a national code

Ref country code: FR

Ref legal event code: ST

Effective date: 20151130

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: GB

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20150306

Ref country code: CH

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20150331

Ref country code: LI

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20150331

Ref country code: DE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20151001

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: FR

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20150331

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MC

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20140331

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: NL

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20150401

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20140331